1
|
Connolly BJ, Saxton SN. Recent updates on the influence of iron and magnesium on vascular, renal, and adipose inflammation and possible consequences for hypertension. J Hypertens 2024; 42:1848-1861. [PMID: 39258532 PMCID: PMC11451934 DOI: 10.1097/hjh.0000000000003829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
The inflammatory status of the kidneys, vasculature, and perivascular adipose tissue (PVAT) has a significant influence on blood pressure and hypertension. Numerous micronutrients play an influential role in hypertension-driving inflammatory processes, and recent reports have provided bases for potential targeted modulation of these micronutrients to reduce hypertension. Iron overload in adipose tissue macrophages and adipocytes engenders an inflammatory environment and may contribute to impaired anticontractile signalling, and thus a treatment such as chelation therapy may hold a key to reducing blood pressure. Similarly, magnesium intake has proven to greatly influence inflammatory signalling and concurrent hypertension in both healthy animals and in a model for chronic kidney disease, demonstrating its potential clinical utility. These findings highlight the importance of further research to determine the efficacy of micronutrient-targeted treatments for the amelioration of hypertension and their potential translation into clinical application.
Collapse
Affiliation(s)
- Benjamin J Connolly
- Divison of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
2
|
Cheng XC, Tong WZ, Rui W, Feng Z, Shuai H, Zhe W. Single-cell sequencing technology in skin wound healing. BURNS & TRAUMA 2024; 12:tkae043. [PMID: 39445224 PMCID: PMC11497848 DOI: 10.1093/burnst/tkae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 10/25/2024]
Abstract
Skin wound healing is a complicated biological process that mainly occurs in response to injury, burns, or diabetic ulcers. It can also be triggered by other conditions such as dermatitis and melanoma-induced skin cancer. Delayed healing or non-healing after skin injury presents an important clinical issue; therefore, further explorations into the occurrence and development of wound healing at the cellular and molecular levels are necessary. Single-cell sequencing (SCS) is used to sequence and analyze the genetic messages of a single cell. Furthermore, SCS can accurately detect cell expression and gene sequences. The use of SCS technology has resulted in the emergence of new concepts pertaining to wound healing, making it an important tool for studying the relevant mechanisms and developing treatment strategies. This article discusses the application value of SCS technology, the effects of the latest research on skin wound healing, and the value of SCS technology in clinical applications. Using SCS to determine potential biomarkers for wound repair will serve to accelerate wound healing, reduce scar formation, optimize drug delivery, and facilitate personalized treatments.
Collapse
Affiliation(s)
- Xu Cheng Cheng
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Wang Zi Tong
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Wang Rui
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Zhao Feng
- Department of Stem Cells and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang 110013, China
| | - Hou Shuai
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Wang Zhe
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| |
Collapse
|
3
|
Li MK, Xing C, Ma LQ. Integrative bioinformatics analysis to screen key genes and signalling pathways related to ferroptosis in obesity. Adipocyte 2023; 12:2264442. [PMID: 37878496 PMCID: PMC10601513 DOI: 10.1080/21623945.2023.2264442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/18/2023] [Indexed: 10/27/2023] Open
Abstract
Ferroptosis is closely associated with the development of disease in the body. However, there are few studies on ferroptosis-related genes (FRGs) in obesity. Therefore, key genes and signalling pathways related to ferroptosis in obesity were screened. Briefly, the RNA sequencing data of obesity and the non-obesity human samples and 259 FRGs were downloaded from GEO database and FerrDb database, respectively. The obesity-related module genes were firstly screened by weighted gene co-expression network analysis (WGCNA) and crossed with differentially expressed genes (DEGs) of obesity/normal samples and FRGs to obtain obesity-ferroptosis related (OFR) DEGs. Then, key genes were screened by PPI network. Next, the correlation of key genes and differential immune cells between obesity and normal samples were further explored by immune infiltration analysis. Finally, microRNA (miRNA)-messenger RNA (mRNA), transcription factor (TF)-mRNA networks and drug-gene interaction networks were constructed. As a result, 17 OFR DEGs were obtained, which mainly participated in processes such as lipid metabolism or adipocyte differentiation. The 4 key genes, STAT3, IL-6, PTGS2, and VEGFA, constituted the network. M2 macrophages, T cells CD8, mast cells activated, and T cells CD4 memory resting had significant differences between obesity and normal samples. Moreover, 51 miRNAs and 164 drugs were predicted for 4 key genes. All in all, this study has screened 4 FRGs, including IL-6, VEGFA, STAT3, and PTGS2, in obesity patients.
Collapse
Affiliation(s)
- Ming-Ke Li
- Digestive Department, The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chang Xing
- Pediatric Hematology and Digestive Department, Qu Jing Maternal and Child Health-care Hospital, Qujing, China
| | - Lan-Qing Ma
- Digestive Department, The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| |
Collapse
|
4
|
Faradina A, Tung YT, Chen SH, Liao YC, Chou MJ, Teng IC, Lin WL, Wang CC, Sheu MT, Chou PY, Shih CK, Skalny AV, Tinkov AA, Chang JS. Djulis Hull Enhances the Efficacy of Ferric Citrate Supplementation via Restoring Normal Iron Efflux through the IL-6-Hepcidin-Ferroportin Pathway in High-Fat-Diet-Induced Obese Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16691-16701. [PMID: 37877289 DOI: 10.1021/acs.jafc.3c02826] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Obesity-related functional iron disorder remains a major nutritional challenge. We evaluated the effects of djulis hull (DH) on iron metabolism in 50% high-fat-diet-induced obese rats supplemented with ferric citrate (2 g iron/kg diet) for 12 weeks. DH supplementation (5, 10, 15% dry weight/kg diet) significantly increased serum and hepatic iron but decreased appetite hormones, body weight, hepcidin, and liver inflammation (all p < 0.05). The Spearman correlation showed that appetite hormones were negatively associated with iron but positively correlated with liver hepcidin (all p < 0.05). A Western blot analysis showed that DH significantly downregulated hepatic hepcidin through the IL-6-JAK-STAT3 and enhanced ferroportin (Fpn) via the Keap1-Nrf2 and PHD2-HIF-2α. An in vitro study revealed that major bioactive compounds of DH, hexacosanol, and squalene suppressed LPS-induced IL-6 and hepcidin but enhanced Fpn expression in activated THP-1 cells. In conclusion, DH may exert nutraceutical properties for the treatment of functional iron disorder and restoration of iron efflux may have beneficial effects on weight control.
Collapse
Affiliation(s)
- Amelia Faradina
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Seu-Hwa Chen
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Chi Liao
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Meng-Jung Chou
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - I-Chun Teng
- Department of Nutritional Services, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Wen-Ling Lin
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Ching-Chiung Wang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Po-Yu Chou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Kuang Shih
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Anatoly V Skalny
- Center for Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Yaroslavl State University, 150001 Yaroslavl, Russia
| | - Alexey A Tinkov
- Center for Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Yaroslavl State University, 150001 Yaroslavl, Russia
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Chinese Taipei Society for the Study of Obesity, CTSSO, Taipei 110, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
5
|
Sprenkle NT, Winn NC, Bunn KE, Zhao Y, Park DJ, Giese BG, Karijolich JJ, Ansel KM, Serezani CH, Hasty AH, Pua HH. The miR-23-27-24 clusters drive lipid-associated macrophage proliferation in obese adipose tissue. Cell Rep 2023; 42:112928. [PMID: 37542720 PMCID: PMC10712211 DOI: 10.1016/j.celrep.2023.112928] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 08/07/2023] Open
Abstract
Identifying molecular circuits that control adipose tissue macrophage (ATM) function is necessary to understand how ATMs contribute to tissue homeostasis and obesity-induced insulin resistance. In this study, we find that mice with a myeloid-specific knockout of the miR-23-27-24 clusters of microRNAs (miRNAs) gain less weight on a high-fat diet but exhibit worsened glucose and insulin tolerance. Analysis of ATMs from these mice shows selectively reduced numbers and proliferation of a recently reported subset of lipid-associated CD9+Trem2+ ATMs (lipid-associated macrophages [LAMs]). Leveraging the role of miRNAs to control networks of genes, we use RNA sequencing (RNA-seq), functional screens, and biochemical assays to identify candidate target transcripts that regulate proliferation-associated signaling. We determine that miR-23 directly targets the mRNA of Eif4ebp2, a gene that restricts protein synthesis and proliferation in macrophages. Altogether, our study demonstrates that control of proliferation of a protective subset of LAMs by noncoding RNAs contributes to protection against diet-induced obesity metabolic dysfunction.
Collapse
Affiliation(s)
- Neil T Sprenkle
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kaitlyn E Bunn
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yang Zhao
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Deborah J Park
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brenna G Giese
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John J Karijolich
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanerbilt-Ingram Cancer Center, Nashville, TN, USA
| | - K Mark Ansel
- Department of Microbiology and Immunology and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - C Henrique Serezani
- Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
6
|
Phan A, Hage M, Zaharia R, Vigan M, Coursault S, Wilson S, Gabali E, Foussier L, Vychnevskaia K, Raffin-Sanson ML, Bretault M. Nutritional Status of Vegetarian Patients Before and After Bariatric Surgery: a Monocentric Retrospective Observational Case-Control Study. Obes Surg 2023; 33:1356-1365. [PMID: 36991254 DOI: 10.1007/s11695-023-06538-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 03/31/2023]
Abstract
The obesity pandemic is associated with an increasing number of bariatric surgeries which allow improvement in obesity-related comorbidities and life expectancy but potentially induce nutritional deficiencies. Vegetarianism becomes more and more popular and exposes as well to vitamin and micronutrient deficiencies. Only one study has explored the impact of vegetarianism on the preoperative nutritional status of eligible patients for bariatric surgery, but none in postoperative care. MATERIALS AND METHODS We conducted a retrospective case-control study in our cohort of bariatric patients, matching 5 omnivores for each vegetarian. We compared their biological profile regarding vitamin and micronutrient blood levels before and 3, 6, 12, and 30 months after surgery. RESULTS We included 7 vegetarians including 4 lacto-ovo-vegetarians (57%), 2 lacto-vegetarians (29%), and one lacto-ovo-pesco-vegetarian (14%). Three years after surgery with equivalent daily standard vitamin supplementation, the two groups showed a similar biological profile including blood levels of ferritin (p = 0.6), vitamin B1 (p = 0.1), and B12 (p = 0.7), while the total median weight loss at 3 years was comparable (39.1% [27.0-46.6] in vegetarians vs 35.7% [10.5-46.5] in omnivores, p = 0.8). We observed no significant difference between vegetarians and omnivores before surgery regarding comorbidities and nutritional status. CONCLUSION It seems that, after bariatric surgery, vegetarian patients taking a standard vitamin supplementation do not show an increased risk of nutritional deficiencies compared to omnivores. However, a larger study with a longer follow-up is needed to confirm these data, including an evaluation of different types of vegetarianism such as veganism.
Collapse
Affiliation(s)
- Aurélie Phan
- Department of Nutrition, European Georges Pompidou Hospital, APHP, Paris, France
| | - Mirella Hage
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Ramona Zaharia
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Marie Vigan
- Clinical Research Unit, AP-HP, Paris-Saclay, Hôpital Ambroise Pare, Boulogne-Billancourt, France
| | - Séverine Coursault
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Sandy Wilson
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Elodie Gabali
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Loic Foussier
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Karina Vychnevskaia
- Department of Surgery and Oncology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Marie-Laure Raffin-Sanson
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France
| | - Marion Bretault
- Department of Endocrinology-Nutrition, Ambroise Paré Hospital, AP-HP, EA4340 Research Unit, Université de Versailles Saint-Quentin-en-Yvelines, University Paris-Saclay, 92100, Boulogne-Billancourt, France.
| |
Collapse
|
7
|
Nassar AY, Meligy FY, Abd-Allah GM, Khallil WA, Sayed GA, Hanna RT, Nassar GA, Bakkar SM. Oral acetylated whey peptides (AWP) as a potent antioxidant, anti-inflammatory, and chelating agent in iron-overloaded rats' spleen. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
8
|
Abstract
High iron is a risk factor for type 2 diabetes mellitus (T2DM) and affects most of its cardinal features: decreased insulin secretion, insulin resistance, and increased hepatic gluconeogenesis. This is true across the normal range of tissue iron levels and in pathologic iron overload. Because of iron's central role in metabolic processes (e.g., fuel oxidation) and metabolic regulation (e.g., hypoxia sensing), iron levels participate in determining metabolic rates, gluconeogenesis, fuel choice, insulin action, and adipocyte phenotype. The risk of diabetes related to iron is evident in most or all tissues that determine diabetes phenotypes, with the adipocyte, beta cell, and liver playing central roles. Molecular mechanisms for these effects are diverse, although there may be integrative pathways at play. Elucidating these pathways has implications not only for diabetes prevention and treatment, but also for the pathogenesis of other diseases that are, like T2DM, associated with aging, nutrition, and iron.
Collapse
Affiliation(s)
- Alexandria V Harrison
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
| | - Felipe Ramos Lorenzo
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
- Department of Veterans Affairs, W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina, USA
| | - Donald A McClain
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
- Department of Veterans Affairs, W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina, USA
| |
Collapse
|
9
|
Hagarty-Waite KA, Totten MS, Pierce M, Armah SM, Erikson KM. Influence of Sex and Strain on Hepatic and Adipose Tissue Trace Element Concentrations and Gene Expression in C57BL/6J and DBA/2J High Fat Diet Models. Int J Mol Sci 2022; 23:ijms232213778. [PMID: 36430257 PMCID: PMC9697485 DOI: 10.3390/ijms232213778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The objective of this study was to determine the influence of sex and strain on the dysregulation of trace element concentration and associative gene expression due to diet induced obesity in adipose tissue and the liver. Male and female C57BL/6J (B6J) and DBA/2J (D2J) were randomly assigned to a normal-fat diet (NFD) containing 10% kcal fat/g or a mineral-matched high-fat diet (HFD) containing 60% kcal fat/g for 16 weeks. Liver and adipose tissue were assessed for copper, iron, manganese, and zinc concentrations and related changes in gene expression. Notable findings include three-way interactions of diet, sex, and strain amongst adipose tissue iron concentrations (p = 0.005), adipose hepcidin expression (p = 0.007), and hepatic iron regulatory protein (IRP) expression (p = 0.012). Cd11c to Cd163 ratio was increased in adipose tissue due to HFD amongst all biological groups except B6J females, for which tissue iron concentrations were reduced due to HFD (p = 0.002). Liver divalent metal transporter 1 (DMT-1) expression was increased due to HFD amongst B6J males (p < 0.005) and females (p < 0.004), which coincides with the reduction in hepatic iron concentrations found in these biological groups (p < 0.001). Sex, strain, and diet affected trace element concentration, the expression of genes that regulate trace element homeostasis, and the expression of macrophages that contribute to tissue iron-handling in adipose tissue. These findings suggest that sex and strain may be key factors that influence the adaptive capacity of iron mismanagement in adipose tissue and its subsequent consequences, such as insulin resistance.
Collapse
Affiliation(s)
| | - Melissa S. Totten
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA
- Department of Chemistry and Physics, Salem College, Winston-Salem, NC 27101, USA
| | - Matthew Pierce
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA
| | - Seth M. Armah
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA
| | - Keith M. Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA
- Correspondence:
| |
Collapse
|
10
|
Elumalai S, Karunakaran U, Moon JS, Won KC. Ferroptosis Signaling in Pancreatic β-Cells: Novel Insights & Therapeutic Targeting. Int J Mol Sci 2022; 23:13679. [PMID: 36430158 PMCID: PMC9690757 DOI: 10.3390/ijms232213679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022] Open
Abstract
Metabolic stress impairs pancreatic β-cell survival and function in diabetes. Although the pathophysiology of metabolic stress is complex, aberrant tissue damage and β-cell death are brought on by an imbalance in redox equilibrium due to insufficient levels of endogenous antioxidant expression in β-cells. The vulnerability of β-cells to oxidative damage caused by iron accumulation has been linked to contributory β-cell ferroptotic-like malfunction under diabetogenic settings. Here, we take into account recent findings on how iron metabolism contributes to the deregulation of the redox response in diabetic conditions as well as the ferroptotic-like malfunction in the pancreatic β-cells, which may offer insights for deciphering the pathomechanisms and formulating plans for the treatment or prevention of metabolic stress brought on by β-cell failure.
Collapse
Affiliation(s)
- Suma Elumalai
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
| | - Udayakumar Karunakaran
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
| | - Jun-Sung Moon
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Kyu-Chang Won
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
11
|
Chaib M, Hafeez BB, Mandil H, Daria D, Pingili AK, Kumari S, Sikander M, Kashyap VK, Chen GY, Anning E, Tripathi MK, Khan S, Behrman S, Yallapu MM, Jaggi M, Makowski L, Chauhan SC. Reprogramming of pancreatic adenocarcinoma immunosurveillance by a microbial probiotic siderophore. Commun Biol 2022; 5:1181. [PMID: 36333531 PMCID: PMC9636404 DOI: 10.1038/s42003-022-04102-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
There is increasing evidence suggesting the role of microbiome alterations in relation to pancreatic adenocarcinoma and tumor immune functionality. However, molecular mechanisms of the interplay between microbiome signatures and/or their metabolites in pancreatic tumor immunosurveillance are not well understood. We have identified that a probiotic strain (Lactobacillus casei) derived siderophore (ferrichrome) efficiently reprograms tumor-associated macrophages (TAMs) and increases CD8 + T cell infiltration into tumors that paralleled a marked reduction in tumor burden in a syngeneic mouse model of pancreatic cancer. Interestingly, this altered immune response improved anti-PD-L1 therapy that suggests promise of a novel combination (ferrichrome and immune checkpoint inhibitors) therapy for pancreatic cancer treatment. Mechanistically, ferrichrome induced TAMs polarization via activation of the TLR4 pathway that represses the expression of iron export protein ferroportin (FPN1) in macrophages. This study describes a novel probiotic based molecular mechanism that can effectively induce anti-tumor immunosurveillance and improve immune checkpoint inhibitors therapy response in pancreatic cancer.
Collapse
Affiliation(s)
- Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Bilal B Hafeez
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
| | - Hassan Mandil
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Deidre Daria
- Department of Microbiology, Immunology and Biochemistry, Memphis, TN, 38163, USA
| | - Ajeeth K Pingili
- Division of Hematology Oncology, Department of Medicine, Memphis, TN, 38163, USA
| | - Sonam Kumari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Mohammed Sikander
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Guo-Yun Chen
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, Memphis, TN, 38163, USA
| | - Emmanuel Anning
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Manish K Tripathi
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Sheema Khan
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | | | - Murali M Yallapu
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Liza Makowski
- Department of Medicine, Division of Hematology and Oncology and the UTHSC Center for Cancer Research, Memphis, TN, 38103, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
| |
Collapse
|
12
|
Joffin N, Gliniak CM, Funcke JB, Paschoal VA, Crewe C, Chen S, Gordillo R, Kusminski CM, Oh DY, Geldenhuys WJ, Scherer PE. Adipose tissue macrophages exert systemic metabolic control by manipulating local iron concentrations. Nat Metab 2022; 4:1474-1494. [PMID: 36329217 DOI: 10.1038/s42255-022-00664-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Abstract
Iron is essential to many fundamental biological processes, but its cellular compartmentalization and concentration must be tightly controlled. Although iron overload can contribute to obesity-associated metabolic deterioration, the subcellular localization and accumulation of iron in adipose tissue macrophages is largely unknown. Here, we show that macrophage mitochondrial iron levels control systemic metabolism in male mice by altering adipocyte iron concentrations. Using various transgenic mouse models to manipulate the macrophage mitochondrial matrix iron content in an inducible fashion, we demonstrate that lowering macrophage mitochondrial matrix iron increases numbers of M2-like macrophages in adipose tissue, lowers iron levels in adipocytes, attenuates inflammation and protects from high-fat-diet-induced metabolic deterioration. Conversely, elevating macrophage mitochondrial matrix iron increases M1-like macrophages and iron levels in adipocytes, exacerbates inflammation and worsens high-fat-diet-induced metabolic dysfunction. These phenotypes are robustly reproduced by transplantation of a small amount of fat from transgenic to wild-type mice. Taken together, we identify macrophage mitochondrial iron levels as a crucial determinant of systemic metabolic homeostasis in mice.
Collapse
Affiliation(s)
- Nolwenn Joffin
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christy M Gliniak
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jan-Bernd Funcke
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vivian A Paschoal
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Clair Crewe
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, Washington University, St. Louis, MO, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Da Young Oh
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
13
|
Ameka M, Hasty AH. Paying the Iron Price: Liver Iron Homeostasis and Metabolic Disease. Compr Physiol 2022; 12:3641-3663. [PMID: 35766833 PMCID: PMC10155403 DOI: 10.1002/cphy.c210039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Iron is an essential metal element whose bioavailability is tightly regulated. Under normal conditions, systemic and cellular iron homeostases are synchronized for optimal function, based on the needs of each system. During metabolic dysfunction, this synchrony is lost, and markers of systemic iron homeostasis are no longer coupled to the iron status of key metabolic organs such as the liver and adipose tissue. The effects of dysmetabolic iron overload syndrome in the liver have been tied to hepatic insulin resistance, nonalcoholic fatty liver disease, and nonalcoholic steatohepatitis. While the existence of a relationship between iron dysregulation and metabolic dysfunction has long been acknowledged, identifying correlative relationships is complicated by the prognostic reliance on systemic measures of iron homeostasis. What is lacking and perhaps more informative is an understanding of how cellular iron homeostasis changes with metabolic dysfunction. This article explores bidirectional relationships between different proteins involved in iron homeostasis and metabolic dysfunction in the liver. © 2022 American Physiological Society. Compr Physiol 12:3641-3663, 2022.
Collapse
Affiliation(s)
- Magdalene Ameka
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Abstract
Adipose tissue is a complex dynamic organ with whole-body immunometabolic influence. Much of the work into understanding the role of immune cells in adipose tissue has been in the context of obesity. These investigations have also uncovered a range of typical (immune) and non-typical functions exerted by adipose tissue leukocytes. Here we provide an overview of the adipose tissue immune system, including its role as an immune reservoir in the whole-body response to infection and as a site of parasitic and viral infections. We also describe the functional roles of specialized immunological structures found within adipose tissue. However, our main focus is on the recently discovered 'non-immune' functions of adipose tissue immune cells, which include the regulation of adipocyte homeostasis, as well as responses to changing nutrient status and body temperature. In doing so, we outline the therapeutic potential of the adipose tissue immune system in health and disease.
Collapse
|
15
|
Mao H, Dou W, Chen K, Wang X, Wang X, Guo Y, Zhang C. Evaluating iron deposition in gray matter nuclei of patients with unilateral middle cerebral artery stenosis using quantitative susceptibility mapping. Neuroimage Clin 2022; 34:103021. [PMID: 35500369 PMCID: PMC9065429 DOI: 10.1016/j.nicl.2022.103021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 03/17/2022] [Accepted: 04/23/2022] [Indexed: 11/18/2022]
Abstract
Iron mediated oxidative stress is involved in the process of brain injury after long-term ischemia. While increased iron deposition in the affected brain regions was observed in animal models of ischemic stroke, potential changes in the brain iron content in clinical patients with cerebral ischemia remain unclear. Quantitative susceptibility mapping (QSM), a non-invasive magnetic resonance imaging technique, can be used to evaluate iron content in the gray matter (GM) nuclei reliably. In this study, we aimed to quantitatively evaluate iron content changes in GM nuclei of patients with long-term unilateral middle cerebral artery (MCA) stenosis/occlusion-related cerebral ischemia using QSM. Forty-six unilateral MCA stenosis/occlusion patients and 38 age-, sex- and education-matched healthy controls underwent QSM. Clinical variables of history of hypertension, diabetes, hyperlipidemia, hyperhomocysteinemia, smoking, and drinking in all patients were evaluated. The iron-related susceptibility of GM nucleus subregions, including the bilateral caudate nucleus (CN), putamen (PU), globus pallidus (GP), thalamus, substantia nigra (SN), red nucleus, and dentate nucleus, was assessed. Susceptibility was compared between the bilateral GM nuclei in patients and controls. Receiver operating characteristic curve analysis was used to evaluate the efficacy of QSM susceptibility in distinguishing patients with unilateral MCA stenosis/occlusion from healthy controls. Multiple linear regression analysis was used to evaluate the relationship between ipsilateral susceptibility levels and clinical variables. Except for the CN, the susceptibility in most bilateral GM nucleus subregions was comparable in healthy controls, whereas for patients with unilateral MCA stenosis/occlusion, the ipsilateral PU, GP, and SN exhibited significantly higher susceptibility than the contralateral side (all P < 0.05). Compared with controls, susceptibility of the ipsilateral PU, GP, and SN and of contralateral PU in patients were significantly increased (all P < 0.05). The area under the curve (AUC) was greater for the ipsilateral PU than for the GP and SN (AUC = 0.773, 0.662 and 0.681; all P < 0.05). Multiple linear regression analysis showed that the increased susceptibility of the ipsilateral PU was significantly associated with hypertension, of the ipsilateral GP associated with smoking, and of the ipsilateral SN associated with diabetes (all P < 0.05). Our findings provide support for abnormal iron accumulation in the GM nuclei after chronic MCA stenosis/occlusion and its correlation with some cerebrovascular disease risk factors. Therefore, iron deposition in the GM nuclei, as measured by QSM, may be a potential biomarker for long-term cerebral ischemia.
Collapse
Affiliation(s)
- Huimin Mao
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China; Shandong First Medical University, Jinan, Shandong Province 250000, China
| | - Weiqiang Dou
- MR Research, GE Healthcare, Beijing 10076, China
| | - Kunjian Chen
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China; Shandong First Medical University, Jinan, Shandong Province 250000, China
| | - Xinyu Wang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China; Shandong First Medical University, Jinan, Shandong Province 250000, China
| | - Xinyi Wang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China.
| | - Yu Guo
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China; Shandong First Medical University, Jinan, Shandong Province 250000, China
| | - Chao Zhang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China
| |
Collapse
|
16
|
Impact of microRNA Regulated Macrophage Actions on Adipose Tissue Function in Obesity. Cells 2022; 11:cells11081336. [PMID: 35456015 PMCID: PMC9024513 DOI: 10.3390/cells11081336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity-induced adipose tissue dysfunction is bolstered by chronic, low-grade inflammation and impairs systemic metabolic health. Adipose tissue macrophages (ATMs) perpetuate local inflammation but are crucial to adipose tissue homeostasis, exerting heterogeneous, niche-specific functions. Diversified macrophage actions are shaped through finely regulated factors, including microRNAs, which post-transcriptionally alter macrophage activation. Numerous studies have highlighted microRNAs’ importance to immune function and potential as inflammation-modulatory. This review summarizes current knowledge of regulatory networks governed by microRNAs in ATMs in white adipose tissue under obesity stress.
Collapse
|
17
|
Chen H, Sun L, Feng L, Yin Y, Zhang W. Role of Innate lymphoid Cells in Obesity and Insulin Resistance. Front Endocrinol (Lausanne) 2022; 13:855197. [PMID: 35574038 PMCID: PMC9091334 DOI: 10.3389/fendo.2022.855197] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity, a growing chronic metabolic disease, greatly increases the risk of metabolic syndrome which includes type 2 diabetes, fatty liver and cardiovascular diseases. Obesity-associated metabolic diseases significantly contribute to mortality and reduce life expectancy. Recently, innate lymphoid cells (ILCs) have emerged as crucial regulators of metabolic homeostasis and tissue inflammation. This review focuses on the roles of ILCs in different metabolic tissues, including adipose tissue, liver, pancreas, and intestine. We briefly outline the relationship between obesity, inflammation, and insulin resistance. We then discuss how ILCs in distinct metabolic organs may function to maintain metabolic homeostasis and contribute to obesity and its associated metabolic diseases. The potential of ILCs as the therapeutic target for obesity and insulin resistance is also addressed.
Collapse
Affiliation(s)
- Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lu Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| |
Collapse
|
18
|
Evaluation of the relationship between serum ferritin and insulin resistance and visceral adiposity index (VAI) in women with polycystic ovary syndrome. Eat Weight Disord 2021; 26:1581-1593. [PMID: 32772321 DOI: 10.1007/s40519-020-00980-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
PURPOSE There is a relationship between polycystic ovary syndrome (PCOS) and adipose tissue dysfunction (ADD), but this relationship is not clear. It has been recently shown that iron accumulation in adipose tissue is among the causes of adipose tissue dysfunction. Data on adipose tissue dysfunction in women with PCOS are insufficient. In this study, we aimed to evaluate the relationship between serum ferritin levels (iron accumulation biomarker) and visceral adiposity index (an indicator of adipose tissue dysfunction). METHODS The study is a case-control study. Women with diagnosed PCOS with 2003 Rotterdam Diagnostic Criteria (n = 40) were compared with non-PCOS group (n = 40). In this study, the cholesterol ratios, the homeostatic model evaluation index for insulin resistance (HOMA-IR) and the quantitative insulin sensitivity control index were calculated using biochemical parameters, and the visceral adiposity index (VAI) and the lipid accumulation product (LAP) were calculated using both anthropometric and biochemical parameters. In this study, insulin resistance was evaluated by HOMA-IR and adipose tissue dysfunction was evaluated by VAI index. RESULTS According to the results of this study, women with PCOS have a worse metabolic status than women without PCOS. However, this has been shown only in overweight and obese women, not in women with normal weight. CONCLUSION As a result, the presence of obesity in women with PCOS exacerbates metabolic status. LEVEL OF EVIDENCE Level V, cross-sectional descriptive study.
Collapse
|
19
|
Macrophage metabolic adaptation to heme detoxification involves CO-dependent activation of the pentose phosphate pathway. Blood 2021; 136:1535-1548. [PMID: 32556090 DOI: 10.1182/blood.2020004964] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023] Open
Abstract
Heme is an essential cofactor for numerous cellular functions, but release of free heme during hemolysis results in oxidative tissue damage, vascular dysfunction, and inflammation. Macrophages play a key protective role in heme clearance; however, the mechanisms that regulate metabolic adaptations that are required for effective heme degradation remain unclear. Here we demonstrate that heme loading drives a unique bioenergetic switch in macrophages, which involves a metabolic shift from oxidative phosphorylation toward glucose consumption. Metabolomic and transcriptional analysis of heme-loaded macrophages revealed that glucose is funneled into the pentose phosphate pathway (PPP), which is indispensable for efficient heme detoxification and is required to maintain redox homeostasis. We demonstrate that the metabolic shift to the PPP is controlled by heme oxygenase-dependent generation of carbon monoxide (CO). Finally, we show that PPP upregulation occurs in vivo in organ systems central to heme clearance and that PPP activity correlates with heme levels in mouse sickle cell disease (SCD). Together, our findings demonstrate that metabolic adaptation to heme detoxification in macrophages requires a shift to the PPP that is induced by heme-derived CO, suggesting pharmacologic targeting of macrophage metabolism as a novel therapeutic strategy to improve heme clearance in patients with hemolytic disorders.
Collapse
|
20
|
Alkazemi D, Rahman A, Habra B. Alterations in glutathione redox homeostasis among adolescents with obesity and anemia. Sci Rep 2021; 11:3034. [PMID: 33542364 PMCID: PMC7862436 DOI: 10.1038/s41598-021-82579-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022] Open
Abstract
The reduced (GSH)-to-oxidized (GSSG) glutathione ratio represents a dynamic balance between oxidants and antioxidants. However, redox status in adolescents with obesity and anemia has not been investigated. This study investigated the association of erythrocyte GSH redox status (GSH, GSH:GSSG ratio, and glutathione peroxidase [GPx] activity) with anemia and adiposity in adolescents. This case–control study nested in a cross-sectional study enrolled 524 adolescents (268 boys; 256 girls). The prevalence of anemia in overweight and obesity (OWOB) was 5.2% in boys and 11.7% in girls. The GSH:GSSG ratio and GPx activity were significantly higher in girls than in boys (p < 0.001), in anemic than in non-anemic subjects (p < 0.001), and in OWOB than in normal-weight subjects (p < 0.001). Similarly, significantly higher GSH: GSSG level (p < 0.001) and GPx activity (p < 0.001) were found in subjects with 90th percentile waist circumference than in those with < 90th percentile. GPx and GSH:GSSG were positively associated with anemia after adjusting for age, sex, and body mass index (adjusted odds ratio, adjOR [95% confidence interval, CI] 2.18 [1.44–3.29]) or tertiles (adjOR [95% CI], T3 = 2.49 [1.03–6.01]). A similar association was noted for GSH and GPx. A compensatory increased redox defense mechanism exists in anemia and obesity among adolescents without metabolic disturbances.
Collapse
Affiliation(s)
- Dalal Alkazemi
- Department of Food Science and Nutrition, College of Life Sciences, Kuwait University, AlShadadiyah, Kuwait.
| | - Abdur Rahman
- Department of Food Science and Nutrition, College of Life Sciences, Kuwait University, AlShadadiyah, Kuwait
| | - Banan Habra
- Department of Food Science and Nutrition, College of Life Sciences, Kuwait University, AlShadadiyah, Kuwait
| |
Collapse
|
21
|
Role of Kallikrein 7 in Body Weight and Fat Mass Regulation. Biomedicines 2021; 9:biomedicines9020131. [PMID: 33572949 PMCID: PMC7912635 DOI: 10.3390/biomedicines9020131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 02/08/2023] Open
Abstract
Increased plasma and adipose tissue protease activity is observed in patients with type 2 diabetes and obesity. It has been proposed that specific proteases contribute to the link between obesity, adipose tissue inflammation and metabolic diseases. We have recently shown that ablation of the serine protease kallikrein-related peptidase 7 (Klk7) specifically in adipose tissue preserves systemic insulin sensitivity and protects mice from obesity-related AT inflammation. Here, we investigated whether whole body Klk7 knockout (Klk7-/-) mice develop a phenotype distinct from that caused by reduced Klk7 expression in adipose tissue. Compared to littermate controls, Klk7-/- mice gain less body weight and fat mass both under chow and high fat diet (HFD) feeding, are hyper-responsive to exogenous insulin and exhibit preserved adipose tissue function due to adipocyte hyperplasia and lower inflammation. Klk7-/- mice exhibit increased adipose tissue thermogenesis, which is not related to altered thyroid function. These data strengthen our recently proposed role of Klk7 in the regulation of body weight, energy metabolism, and obesity-associated adipose tissue dysfunction. The protective effects of Klk7 deficiency in obesity are likely linked to a significant limitation of adipocyte hypertrophy. In conclusion, our data indicate potential application of specific KLK7 inhibitors to regulate KLK7 activity in the development of obesity and counteract obesity-associated inflammation and metabolic diseases.
Collapse
|
22
|
Li J, Zhang Q, Zhang N, Guo L. Increased Brain Iron Detection by Voxel-Based Quantitative Susceptibility Mapping in Type 2 Diabetes Mellitus Patients With an Executive Function Decline. Front Neurosci 2021; 14:606182. [PMID: 33519360 PMCID: PMC7843466 DOI: 10.3389/fnins.2020.606182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
Purpose Brain iron accumulation has been suggested as a pathomechanism in patients with type 2 diabetes mellitus (T2DM) with cognitive impairment. This research aims to examine the total-brain pattern of iron accumulation in relation to executive function decline in patients with T2DM by voxel-based quantitative susceptibility mapping (QSM) analysis. Materials and Methods A total of 32 patients with T2DM and 34 age- and sex-matched healthy controls (HCs) were enrolled in this study. All participants underwent brain magnetic resonance examination, and 48 individuals underwent cognitive function assessments. Imaging data were collected with three-dimensional fast low-angle shot sequences to achieve magnitude as well as phase images. Using voxel-based QSM analysis, we compared the voxel-wise susceptibility values of the whole brain among groups and explored whether the susceptibility values had correlations with cognitive data. Results Among the 66 participants, cognitive function was estimated in 23 patients with T2DM (11 males and 12 females; average age, 64.65 ± 8.44 years) and 25 HCs (13 males and 12 females; average age, 61.20 ± 7.62 years). T2DM patients exhibited significantly (t = 4.288, P < 0.001) lower Montreal Cognitive Assessment (MoCA) scores [T2DM, 27 (27, 28); HCs, 29 (28, 29); normal standard ≥ 26)] and higher Trail-making Test (TMT)-A/TMT-B scores [71 (51, 100)/185 (149, 260)] than HCs [53 (36.5, 63.5)/150 (103, 172.5)] (Z = 2.612, P = 0.009; Z = 2.797, P = 0.005). Subjects with T2DM showed significantly higher susceptibility values than HCs in the caudate/putamen/pallidum, frontal inferior triangular gyrus, and precentral gyrus on the right hemisphere. In contrast (HC > T2DM), no region showed a significant difference in susceptibility values between the groups. The correlation analysis between susceptibility values and cognitive function scores was tested by voxel-based susceptibility value with sex and age as covariates. After multiple comparison correction, in T2DM patients, the left thalamus showed a significant relationship with TMT-A (R 2 = 0.53, P = 0.001). The right thalamus and left thalamus showed a significant relationship with TMT-B (R 2 = 0.35, P = 0.019; and R 2 = 0.38, P = 0.017, respectively). In HCs, the cluster of right precentral/middle frontal gyrus/inferior frontal gyrus/inferior triangular gyrus showed a significant relationship with TMT-B (R 2 = 0.59, P = 0.010). No relationship was found between the susceptibility values with MoCA in the brain region in both two groups. Conclusion Patients with T2DM presented declined cognitive assessments and elevated iron deposition in the striatum and frontal lobe, suggesting that executive function decline in T2DM might be associated with the cerebral iron burden and that changes in susceptibility values may represent a latent quantitative imaging marker for early assessment of cognitive decline in patients with T2DM.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qihao Zhang
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Nan Zhang
- Shandong Medical Imaging Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lingfei Guo
- Shandong Medical Imaging Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
23
|
Salcedo-Tacuma D, Parales-Giron J, Prom C, Chirivi M, Laguna J, Lock AL, Contreras GA. Transcriptomic profiling of adipose tissue inflammation, remodeling, and lipid metabolism in periparturient dairy cows (Bos taurus). BMC Genomics 2020; 21:824. [PMID: 33228532 PMCID: PMC7686742 DOI: 10.1186/s12864-020-07235-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/17/2020] [Indexed: 12/30/2022] Open
Abstract
Background Periparturient cows release fatty acid reserves from adipose tissue (AT) through lipolysis in response to the negative energy balance induced by physiological changes related to parturition and the onset of lactation. However, lipolysis causes inflammation and structural remodeling in AT that in excess predisposes cows to disease. The objective of this study was to determine the effects of the periparturient period on the transcriptomic profile of AT using NGS RNAseq. Results Subcutaneous AT samples were collected from Holstein cows (n = 12) at 11 ± 3.6 d before calving date (PreP) and at 6 ± 1d (PP1) and 13 ± 1.4d (PP2) after parturition. Differential expression analyses showed 1946 and 1524 DEG at PP1 and PP2, respectively, compared to PreP. Functional Enrichment Analysis revealed functions grouped in categories such as lipid metabolism, molecular transport, energy production, inflammation, and free radical scavenging to be affected by parturition and the onset of lactation (FDR < 0.05). Inflammation related genes such as TLR4 and IL6 were categorized as upstream lipolysis triggers. In contrast, FASN, ELOVL6, ACLS1, and THRSP were identified as upstream inhibitors of lipid synthesis. Complement (C3), CXCL2, and HMOX1 were defined as links between inflammatory pathways and those involved in the generation of reactive oxygen species. Conclusions Results offer a comprehensive characterization of gene expression dynamics in periparturient AT, identify upstream regulators of AT function, and demonstrate complex interactions between lipid mobilization, inflammation, extracellular matrix remodeling, and redox signaling in the adipose organ. Supplementary Information Supplementary information accompanies this paper at 10.1186/s12864-020-07235-0.
Collapse
Affiliation(s)
- David Salcedo-Tacuma
- Department of Physiology, College of Natural Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Jair Parales-Giron
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, 48824, USA
| | - Crystal Prom
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, 48824, USA
| | - Miguel Chirivi
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Juliana Laguna
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, 48824, USA.,Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Adam L Lock
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, 48824, USA
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
24
|
Fillebeen C, Lam NH, Chow S, Botta A, Sweeney G, Pantopoulos K. Regulatory Connections between Iron and Glucose Metabolism. Int J Mol Sci 2020; 21:ijms21207773. [PMID: 33096618 PMCID: PMC7589414 DOI: 10.3390/ijms21207773] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023] Open
Abstract
Iron is essential for energy metabolism, and states of iron deficiency or excess are detrimental for organisms and cells. Therefore, iron and carbohydrate metabolism are tightly regulated. Serum iron and glucose levels are subjected to hormonal regulation by hepcidin and insulin, respectively. Hepcidin is a liver-derived peptide hormone that inactivates the iron exporter ferroportin in target cells, thereby limiting iron efflux to the bloodstream. Insulin is a protein hormone secreted from pancreatic β-cells that stimulates glucose uptake and metabolism via insulin receptor signaling. There is increasing evidence that systemic, but also cellular iron and glucose metabolic pathways are interconnected. This review article presents relevant data derived primarily from mouse models and biochemical studies. In addition, it discusses iron and glucose metabolism in the context of human disease.
Collapse
Affiliation(s)
- Carine Fillebeen
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC H3Y 1P3, Canada;
| | - Nhat Hung Lam
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Samantha Chow
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Amy Botta
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC H3Y 1P3, Canada;
- Correspondence: ; Tel.: +1-514-340-8260 (ext. 25293)
| |
Collapse
|
25
|
Faria SS, Corrêa LH, Heyn GS, de Sant'Ana LP, Almeida RDN, Magalhães KG. Obesity and Breast Cancer: The Role of Crown-Like Structures in Breast Adipose Tissue in Tumor Progression, Prognosis, and Therapy. J Breast Cancer 2020; 23:233-245. [PMID: 32595986 PMCID: PMC7311368 DOI: 10.4048/jbc.2020.23.e35] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is associated with increased risk and aggressiveness of many types of cancer. Women with obesity and breast cancer are more likely to be diagnosed with larger and higher-grade tumors and have higher incidence of metastases than lean individuals. Increasing evidence indicates that obesity includes systemic, chronic low-grade inflammation, and that adipose tissue can act as an important endocrine site, secreting a variety of substances that may regulate inflammation, immune response, and cancer predisposition. Obesity-associated inflammation appears to be initially mediated by macrophage infiltration into adipose tissue. Macrophages can surround damaged or necrotic adipocytes, forming "crown-like" structures (CLS). CLS are increased in breast adipose tissue from breast cancer patients and are more abundant in patients with obesity conditions. Moreover, the CLS index-ratio from individuals with obesity seems to influence breast cancer recurrence rates and survival. In this review, we discuss the most recent cellular and molecular mechanisms involved in CLS establishment in the white adipose tissue of women with obesity and their implications for breast cancer biology. We also explain how CLS influence the tumor microenvironment and affect breast cancer behavior. Targeting breast adipose tissue CLS can be a crucial therapeutic tool in cancer treatment, especially in patients with obesity.
Collapse
Affiliation(s)
- Sara Socorro Faria
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Gabriella Simões Heyn
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Lívia Pimentel de Sant'Ana
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Raquel das Neves Almeida
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
26
|
Association of Iron Storage Markers with Metabolic Syndrome and Its Components in Chinese Rural 6-12 Years Old Children: The 2010-2012 China National Nutrition and Health Survey. Nutrients 2020; 12:nu12051486. [PMID: 32443740 PMCID: PMC7284848 DOI: 10.3390/nu12051486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 12/28/2022] Open
Abstract
Background: Elevated ferritin, which is often used to represent iron storage, is known to increase the risk of metabolic syndrome (MetS) or its components, but its increase is affected by many factors. Therefore, it is necessary to analyze the relationship between other indicators of iron storage, and MetS and its components in order to fully understand the role of iron in the occurrence and development of these diseases. Although there are many studies to analyze the relationship involved in adults and adolescents, in children there is limited research. In this study, we aim to estimate the association of whole blood iron, ferritin, and total body iron with metabolic syndrome, and especially its components in Chinese rural children aged 6–12 years old. Method: A total of 1333 children aged 6–12 years old were enrolled from the 2010–2012 China National Nutrition and Health Survey in this study. Markers of iron storage (whole blood iron, ferritin, and total body iron (TBI)) and MetS component parameters (waist, blood pressure, high-density lipoprotein cholesterol (HDL-C), triglyceride (TG), and fast glycose) were collected. A multivariate logistic regression analysis was performed to confirm the independent relationship between iron storage markers, and the incident of metabolic syndrome and its components. Results: After adjusting for age, gender, C-reactive protein (CRP), and body mass index (BMI), a negative association was found between whole blood iron, ferritin, and TBI and incidence of reduced HDL-C (odds ratio (OR) = 0.63, 0.49, and 0.57, respectively). The highest tertile of whole blood iron increased the risk of the incidence of hyperglycemia (OR = 1.74), while TBI decreased the risk by 61%. No significant association was found between ferritin tertiles and the incidence of hyperglycemia. Conclusion: An iron storage level within the normal range in children is associated with a risk of MetS components, especially in hyperglycemia and reduced HDL-C. The relationship between the three iron indexes and metabolic syndrome and its components is not completely consistent, which suggests that the underlying mechanism is complex and needs to be further explored.
Collapse
|
27
|
Chaib M, Chauhan SC, Makowski L. Friend or Foe? Recent Strategies to Target Myeloid Cells in Cancer. Front Cell Dev Biol 2020; 8:351. [PMID: 32509781 PMCID: PMC7249856 DOI: 10.3389/fcell.2020.00351] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is a complex network of epithelial and stromal cells, wherein stromal components provide support to tumor cells during all stages of tumorigenesis. Among these stromal cell populations are myeloid cells, which are comprised mainly of tumor-associated macrophages (TAM), dendritic cells (DC), myeloid-derived suppressor cells (MDSC), and tumor-associated neutrophils (TAN). Myeloid cells play a major role in tumor growth through nurturing cancer stem cells by providing growth factors and metabolites, increasing angiogenesis, as well as promoting immune evasion through the creation of an immune-suppressive microenvironment. Immunosuppression in the TME is achieved by preventing critical anti-tumor immune responses by natural killer and T cells within the primary tumor and in metastatic niches. Therapeutic success in targeting myeloid cells in malignancies may prove to be an effective strategy to overcome chemotherapy and immunotherapy limitations. Current therapeutic approaches to target myeloid cells in various cancers include inhibition of their recruitment, alteration of function, or functional re-education to an antitumor phenotype to overcome immunosuppression. In this review, we describe strategies to target TAMs and MDSCs, consisting of single agent therapies, nanoparticle-targeted approaches and combination therapies including chemotherapy and immunotherapy. We also summarize recent molecular targets that are specific to myeloid cell populations in the TME, while providing a critical review of the limitations of current strategies aimed at targeting a single subtype of the myeloid cell compartment. The goal of this review is to provide the reader with an understanding of the critical role of myeloid cells in the TME and current therapeutic approaches including ongoing or recently completed clinical trials.
Collapse
Affiliation(s)
- Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Subhash C Chauhan
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Liza Makowski
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Hematology Oncology, Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
28
|
Li J, Zhang Q, Zhang N, Guo L. Increased Brain Iron Deposition in the Putamen in Patients with Type 2 Diabetes Mellitus Detected by Quantitative Susceptibility Mapping. J Diabetes Res 2020; 2020:7242530. [PMID: 33062715 PMCID: PMC7533753 DOI: 10.1155/2020/7242530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/14/2020] [Accepted: 09/11/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The underlying brain structural changes in type 2 diabetes mellitus (T2DM) patients have attracted increasing attention. The insulin-resistant state causes iron overload in neurons and leads to lesions in the central nervous system. Quantitative susceptibility mapping (QSM) can provide a noninvasive quantitative analysis of brain iron deposition. We aimed to compare the difference of brain iron deposition in the gray matter nucleus between T2DM patients and healthy elderly individuals using QSM. METHODS Thirty-two T2DM patients and thirty-two age- and gender-matched healthy controls (HCs) were enrolled in this research. Twenty-three patients and twenty-six HCs underwent cognitive assessments. Brain QSM maps were computed from multiecho GRE data using morphology-enabled dipole inversion with automatic uniform cerebrospinal fluid zero reference algorithm (MEDI+0). ITK-SNAP was used to measure the susceptibility values reflecting the content of iron in the regions of interest (ROIs). RESULTS The study included thirty-two T2DM patients (20 males and 12 females; mean age of 61.09 ± 9.99 years) and 32 HCs (14 males and 18 females; mean age of 59.09 ± 9.77 years). These participants had no significant difference in age or gender (P > 0.05). Twenty-three patients with T2DM (11 males and 12 females; mean age, 64.65 ± 8.44 years) and twenty-six HCs (14 males and 12 females; mean age, 62.30 ± 6.13 years) received an assessment of cognitive function. T2DM patients exhibited an obviously (t = 3.237, P = 0.003) lower Montreal Cognitive Assessment (MoCA) score (26.78 ± 2.35; HCs, 28.42 ± 0.64; normal standard ≥26) and a higher Stroop color-word test (SCWT)-C score [87(65,110); HC, 63(60,76.75), Z = -2.232, P = 0.003] than HCs. The mean susceptibility values in the putamen appeared obviously higher in T2DM patients than in HCs (t = -3.994, P < 0.001). The susceptibility values and cognitive assessment scores showed no obvious association (P > 0.05). However, an obvious correlation was observed between the changes in the susceptibility values in the putamen and the thalamus/dentate nucleus (r = 0.404, P < 0.001; r = 0.423, P < 0.001). CONCLUSION T2DM patients showed increased susceptibility values in the putamen and had declines in executive functions, but the linear association between them was not statistically significant. Changes in susceptibility values in the putamen indicated increased iron deposition and might be used as a quantitative imaging marker of central nervous system injury in T2DM patients. QSM might be able to help probe micro neuronal damage in gray matter and provide information on diabetic encephalopathy.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China. 95 Yongan Road, Xi Cheng District, Beijing 100050, China
| | - Qihao Zhang
- Department of Radiology, Weill Cornell Medical College, New York. 71st E No. 515, 10044 New York, USA
| | - Nan Zhang
- Shandong Medical Imaging Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China. Jing-wu Road No. 324, Jinan, Shandong 250021, China
| | - Lingfei Guo
- Shandong Medical Imaging Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China. Jing-wu Road No. 324, Jinan, Shandong 250021, China
| |
Collapse
|
29
|
|
30
|
Antolic A, Richards EM, Wood CE, Keller-Wood M. A Transcriptomic Model of Postnatal Cardiac Effects of Prenatal Maternal Cortisol Excess in Sheep. Front Physiol 2019; 10:816. [PMID: 31333485 PMCID: PMC6616147 DOI: 10.3389/fphys.2019.00816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/11/2019] [Indexed: 12/25/2022] Open
Abstract
In utero treatment with glucocorticoids have been suggested to reprogram postnatal cardiovascular function and stress responsiveness. However, little is known about the effects of prenatal exposure to the natural corticosteroid, cortisol, on postnatal cardiovascular system or metabolism. We have demonstrated an increased incidence of stillbirth in sheep pregnancies in which there is mild maternal hypercortisolemia caused by infusion of 1 mg/kg/d cortisol. In order to model corticosteroid effects in the neonate, we created a second model in which cortisol was infused for 12 h per day for a daily infusion of 0.5 mg/kg/d. In this model we had previously found that neonatal plasma glucose was increased and plasma insulin was decreased compared to those in the control group, and that neonatal ponderal index and kidney weight were reduced and left ventricular wall thickness was increased in the 2 week old lamb. In this study, we have used transcriptomic modeling to better understand the programming effect of this maternal hypercortisolemia in these hearts. This is a time when both terminal differentiation and a shift in the metabolism of the heart from carbohydrates to lipid oxidation are thought to be complete. The transcriptomic model indicates suppression of genes in pathways for fatty acid and ketone production and upregulation of genes in pathways for angiogenesis in the epicardial adipose fat (EAT). The transcriptomic model indicates that RNA related pathways are overrepresented by downregulated genes, but ubiquitin-mediated proteolysis and protein targeting to the mitochondria are overrepresented by upregulated genes in the intraventricular septum (IVS) and left ventricle (LV). In IVS the AMPK pathway and adipocytokine signaling pathways were also modeled based on overrepresentation by downregulated genes. Peroxisomal activity is modeled as increased in EAT, but decreased in LV and IVS. Our results suggest that pathways for lipids as well as cell proliferation and cardiac remodeling have altered activity postnatally after the in utero cortisol exposure. Together, this model is consistent with the observed increase in cardiac wall thickness at necropsy and altered glucose metabolism observed in vivo, and predicts that in utero exposure to excess maternal cortisol will cause postnatal cardiac hypertrophy and altered responses to oxidative stress.
Collapse
Affiliation(s)
- Andrew Antolic
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Elaine M Richards
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States.,Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
31
|
Echeverría R, Vrhovnik P, Salcedo-Bellido I, Iribarne-Durán LM, Fiket Ž, Dolenec M, Martin-Olmedo P, Olea N, Arrebola JP. Levels and determinants of adipose tissue cadmium concentrations in an adult cohort from Southern Spain. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 670:1028-1036. [PMID: 31018418 DOI: 10.1016/j.scitotenv.2019.03.114] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 06/09/2023]
Abstract
This study was conceived as a first step to evaluate the suitability of adipose tissue cadmium (Cd) concentrations as a biomarker for the assessment of long-term exposure. Specifically, the aim of this work was to explore the socio-demographic, dietary, and lifestyle determinants of adipose tissue Cd concentrations. The study population is a subsample of GraMo cohort. Adipose tissue samples were intraoperatively collected from 226 adult volunteers recruited in two public hospitals from Granada, Spain. Cd concentrations in adipose tissue were analyzed by High-Resolution Inductively Coupled Plasma Mass Spectrometry (HR-ICP-MS). Data on socio-demographic characteristics, lifestyle, diet and health status were collected by face-to-face interviews. Predictors of Cd concentrations were assessed by multivariable linear regression with a stepwise variable selection. We found detectable levels of Cd in the adipose tissue of all the study participants, with a mean concentration (±standard deviation) of 12.66 ± 18.91 μg/kg. Smoking habit at recruitment was associated with increased adipose tissue Cd concentrations (β for smokers = 0.669 p < 0.001; β for former smokers = 0.502, p < 0.001; reference = non-smokers). Age was positively associated with Cd concentrations (β = 0.014, p < 0.001), and men showed lower concentrations than women (β = -0.424, p < 0.001). Obesity, measured as Body Mass Index (BMI), showed an inverse association with Cd concentrations (β = -0.038, p < 0.001). Egg consumption ≥2 portions/week (β = 0.241, p = 0.025) was positively associated with Cd concentrations. Perceived exposure to paints was also positively associated with Cd concentrations. The observed associations with age, smoking habit, BMI, and egg and meat consumption did not substantially change after sex/gender stratification. Our results are consistent with currently-known Cd sources and suggest other potential pathways, which might be population-specific. As a whole, our findings underline the potential relevance of adipose tissue as a biological matrix for exposure characterization to Cd, as well as for the assessment of long-term clinical implications of the exposure, particularly in obesity-related diseases.
Collapse
Affiliation(s)
| | - Petra Vrhovnik
- Slovenian National Building and Civil Engineering Institute (ZAG), Ljubjana, Slovenia
| | - Inmaculada Salcedo-Bellido
- University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, Hospitales Universitarios de Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Luz María Iribarne-Durán
- University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, Hospitales Universitarios de Granada, Spain
| | - Željka Fiket
- Ruđer Bošković Institute, Division for Marine and Environmental Research, Zagreb, Croatia
| | - Matej Dolenec
- University of Ljubljana, Faculty of Natural Sciences and Engineering, Department of Geology, Ljubljana, Slovenia
| | - Piedad Martin-Olmedo
- Instituto de Investigación Biosanitaria de Granada, Hospitales Universitarios de Granada, Spain; Escuela Andaluza de Salud Pública, Granada, Spain
| | - Nicolás Olea
- University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, Hospitales Universitarios de Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Juan Pedro Arrebola
- University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria de Granada, Hospitales Universitarios de Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain.
| |
Collapse
|
32
|
Kizalaite A, Brimiene V, Brimas G, Kiuberis J, Tautkus S, Zarkov A, Kareiva A. Determination of Trace Elements in Adipose Tissue of Obese People by Microwave-Assisted Digestion and Inductively Coupled Plasma Optical Emission Spectrometry. Biol Trace Elem Res 2019; 189:10-17. [PMID: 30032403 DOI: 10.1007/s12011-018-1450-7] [Citation(s) in RCA: 5] [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: 07/03/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022]
Abstract
In the present work, analytical method for the determination of trace elements in adipose tissue by means of inductively coupled plasma optical emission spectrometry (ICP-OES) was developed. Adipose tissue from two groups of the patients with obesity (with and without metabolic syndrome) was investigated. The main aim of this study was to reveal some differences and regularities in concentrations of trace elements in adipose tissue between these two groups of the individuals. Moreover, different types of adipose tissue (subcutaneous, preperitoneal, and visceral) were analyzed separately in order to investigate distribution of metals between these types in different groups of obese people. Al, Ba, Ca, Co, Cu, Cr, Fe, K, Li, Mg, Mn, Na, Ni, Sr, and Zn were selected for the quantitative determination. However, only 6 elements (Na, K, Ca, Fe, Mg, and Zn) were determined in all analyzed samples independently of the type of adipose tissue and presence of metabolic disorder of the patient. Concentrations of Ca, Fe, K, and Na were found to be strongly dependent on the type of adipose tissue. Other elements in terms of detection frequency in adipose tissue samples can be arranged in the following sequence Sr (94.7%), Cr (86.2%), Cu (24.6%), Li (18%), Ba (1.8%), Co (0.05%). The concentrations of Al, Mn, and Ni were found to be lower than limit of detection (LOD) in all analyzed samples. Comparison of metal distribution depending on the type of adipose tissue of people with and without metabolic syndrome is discussed in this work.
Collapse
Affiliation(s)
- Agne Kizalaite
- Institute of Chemistry, Vilnius University, Naugarduko 24, LT-03225, Vilnius, Lithuania
| | - Vilma Brimiene
- Clinic of Gastroenterology, Nephrourology and Surgery, Faculty of Medicine, Center of Abdominal Surgery, Vilnius University, Santariskiu 2, LT-08661, Vilnius, Lithuania
| | - Gintautas Brimas
- Clinic of Gastroenterology, Nephrourology and Surgery, Department of General Surgery, Vilnius University, Šiltnamių 29, LT-04130, Vilnius, Lithuania
| | - Jonas Kiuberis
- Institute of Chemistry, Vilnius University, Naugarduko 24, LT-03225, Vilnius, Lithuania
| | - Stasys Tautkus
- Institute of Chemistry, Vilnius University, Naugarduko 24, LT-03225, Vilnius, Lithuania
| | - Aleksej Zarkov
- Institute of Chemistry, Vilnius University, Naugarduko 24, LT-03225, Vilnius, Lithuania.
| | - Aivaras Kareiva
- Institute of Chemistry, Vilnius University, Naugarduko 24, LT-03225, Vilnius, Lithuania
| |
Collapse
|
33
|
Fakharzadeh S, Kalanaky S, Hafizi M, Nazaran MH, Zardooz H. DIBc, a nanochelating-based nano metal-organic framework, shows anti-diabetic effects in high-fat diet and streptozotocin-induced diabetic rats. Int J Nanomedicine 2019; 14:2145-2156. [PMID: 30988614 PMCID: PMC6443220 DOI: 10.2147/ijn.s196050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aims Despite daily increase in diabetic patients in the world, currently approved medications for this disease, at best, only reduce its progression speed. Using novel technologies is a solution for synthetizing more efficient medicines. In the present study, we evaluated anti-diabetic effects of DIBc, a nano metal–organic framework, which is synthetized based on nanochelating technology. Methods High-fat diet and streptozotocin-induced diabetic rats were treated by DIBc or metformin for 6 weeks. Results DIBc decreased plasma glucose, triglyceride, cholesterol, high-density lipoprotein, and low-density lipoprotein compared with diabetic and metformin groups. In DIBc-treated rats, significant homeostasis model assessment of insulin resistance index, malondialdehyde, and tumor necrosis factor-α decrease was observed. H&E staining showed increased islet number and area in DIBc-treated rats compared with diabetic controls. Conclusion The results showed anti-diabetic effects of nanochelating-based framework. So DIBc, as a nano structure, has the capacity to be evaluated in future studies as a novel anti-diabetic agent.
Collapse
Affiliation(s)
- Saideh Fakharzadeh
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran, .,Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran, .,Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran,
| | - Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran,
| | | | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran, .,Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,
| |
Collapse
|
34
|
Ferritin regulates organismal energy balance and thermogenesis. Mol Metab 2019; 24:64-79. [PMID: 30954544 PMCID: PMC6531837 DOI: 10.1016/j.molmet.2019.03.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE The ferritin heavy/heart chain (FTH) gene encodes the ferroxidase component of the iron (Fe) sequestering ferritin complex, which plays a central role in the regulation of cellular Fe metabolism. Here we tested the hypothesis that ferritin regulates organismal Fe metabolism in a manner that impacts energy balance and thermal homeostasis. METHODS We developed a mouse strain, referred herein as FthR26 fl/fl, expressing a tamoxifen-inducible Cre recombinase under the control of the Rosa26 (R26) promoter and carrying two LoxP (fl) sites: one at the 5'end of the Fth promoter and another the 3' end of the first Fth exon. Tamoxifen administration induces global deletion of Fth in adult FthR26Δ/Δ mice, testing whether FTH is required for maintenance of organismal homeostasis. RESULTS Under standard nutritional Fe supply, Fth deletion in adult FthR26Δ/Δ mice led to a profound deregulation of organismal Fe metabolism, oxidative stress, inflammation, and multi-organ damage, culminating in death. Unexpectedly, Fth deletion was also associated with a profound atrophy of white and brown adipose tissue as well as with collapse of energy expenditure and thermogenesis. This was attributed mechanistically to mitochondrial dysfunction, as assessed in the liver and in adipose tissue. CONCLUSION The FTH component of ferritin acts as a master regulator of organismal Fe homeostasis, coupling nutritional Fe supply to organismal redox homeostasis, energy expenditure and thermoregulation.
Collapse
|
35
|
Bader JE, Enos RT, Velázquez KT, Carson MS, Sougiannis AT, McGuinness OP, Robinson CM, Murphy EA. Repeated clodronate-liposome treatment results in neutrophilia and is not effective in limiting obesity-linked metabolic impairments. Am J Physiol Endocrinol Metab 2019; 316:E358-E372. [PMID: 30576244 PMCID: PMC6415716 DOI: 10.1152/ajpendo.00438.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Depletion of macrophages is thought to be a therapeutic option for obesity-induced inflammation and metabolic dysfunction. However, whether the therapeutic effect is a direct result of reduced macrophage-derived inflammation or secondary to decreases in fat mass is controversial, as macrophage depletion has been shown to disrupt energy homeostasis. This study was designed to determine if macrophage depletion via clodronate-liposome (CLD) treatment could serve as an effective intervention to reduce obesity-driven inflammatory and metabolic impairments independent of changes in energy intake. After 16 wk on a high-fat diet (HFD) or the AIN-76A control (low-fat) diet (LFD) ( n = 30/diet treatment), male C57BL/6J mice were assigned to a CLD- or PBS-liposome treatment ( n = 15/group) for 4 wk. Liposomes were administered biweekly via intraperitoneal injections (8 administrations in total). PBS-liposome-treated groups were pair-fed to their CLD-treated dietary counterparts. Metabolic function was assessed before and after liposome treatment. Adipose tissue, as well as the liver, was investigated for macrophage infiltration and the presence of inflammatory mediators. Additionally, a complete blood count was performed. CLD treatment reduced energy intake. When controlling for energy intake, CLD treatment was unable to regress metabolic dysfunction or nonalcoholic fatty liver disease and impaired adipose tissue insulin action. Moreover, repeated CLD treatment induced neutrophilia and anemia, increased adipose tissue mRNA expression of the proinflammatory cytokines IL-6 and IL-1β, and augmented circulating IL-6 and monocyte chemoattractant protein-1 concentrations ( P < 0.05). This study suggests that repeated intraperitoneal administration of CLD to deplete macrophages attenuates obesity by limiting energy intake. Moreover, after controlling for the benefits of weight loss, the accompanying detrimental side effects limit regular CLD treatment as an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jackie E Bader
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Meredith S Carson
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Alex T Sougiannis
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Cory M Robinson
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University , Morgantown, West Virginia
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| |
Collapse
|
36
|
Ren W, Xia Y, Chen S, Wu G, Bazer FW, Zhou B, Tan B, Zhu G, Deng J, Yin Y. Glutamine Metabolism in Macrophages: A Novel Target for Obesity/Type 2 Diabetes. Adv Nutr 2019; 10:321-330. [PMID: 30753258 PMCID: PMC6416106 DOI: 10.1093/advances/nmy084] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/04/2018] [Accepted: 09/27/2018] [Indexed: 12/23/2022] Open
Abstract
Obesity is a nutritional disorder resulting from a chronic imbalance between energy intake and expenditure. This disease is characterized by inflammation in multiple cell types, including macrophages. M1 macrophage responses are correlated with the progression of obesity or diabetes; therefore, strategies that induce repolarization of macrophages from an M1 to an M2 phenotype may be promising for the prevention of obesity- or diabetes-associated pathology. Glutamine (the most abundant amino acid in the plasma of humans and many other mammals including rats) is effective in inducing polarization of M2 macrophages through the glutamine-UDP-N-acetylglucosamine pathway and α-ketoglutarate produced via glutaminolysis, whereas succinate synthesized via glutamine-dependent anerplerosis or the γ-aminobutyric acid shunt promotes polarization of M1 macrophages. Interestingly, patients with obesity or diabetes show altered glutamine metabolism, including decreases in glutamine and α-ketoglutarate concentrations in serum but increases in succinate concentrations. Thus, manipulation of macrophage polarization through glutamine metabolism may provide a potential target for prevention of obesity- or diabetes-associated pathology.
Collapse
Affiliation(s)
- Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yaoyao Xia
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siyuan Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT
| | - Bie Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Guoqiang Zhu
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Academics Working Station at The First Affiliated Hospital, Changsha Medical University, Changsha, China
| |
Collapse
|
37
|
Stevenson MJ, Uyeda KS, Harder NHO, Heffern MC. Metal-dependent hormone function: the emerging interdisciplinary field of metalloendocrinology. Metallomics 2019; 11:85-110. [PMID: 30270362 PMCID: PMC10249669 DOI: 10.1039/c8mt00221e] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
For over 100 years, there has been an incredible amount of knowledge amassed concerning hormones in the endocrine system and their central role in human health. Hormones represent a diverse group of biomolecules that are released by glands, communicate signals to their target tissue, and are regulated by feedback loops to maintain organism health. Many disease states, such as diabetes and reproductive disorders, stem from misregulation or dysfunction of hormones. Increasing research is illuminating the intricate roles of metal ions in the endocrine system where they may act advantageously in concert with hormones or deleteriously catalyze hormone-associated disease states. As the critical role of metal ions in the endocrine system becomes more apparent, it is increasingly important to untangle the complex mechanisms underlying the connections between inorganic biochemistry and hormone function to understand and control endocrinological phenomena. This tutorial review harmonizes the interdisciplinary fields of endocrinology and inorganic chemistry in the newly-termed field of "metalloendocrinology". We describe examples linking metals to both normal and aberrant hormone function with a focus on highlighting insight to molecular mechanisms. Hormone activities related to both essential metal micronutrients, such as copper, iron, zinc, and calcium, and disruptive nonessential metals, such as lead and cadmium are discussed.
Collapse
Affiliation(s)
- Michael J Stevenson
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
38
|
Kosacka J, Woidt K, Toyka KV, Paeschke S, Klöting N, Bechmann I, Blüher M, Thiery J, Ossmann S, Baum P, Nowicki M. The role of dietary non-heme iron load and peripheral nerve inflammation in the development of peripheral neuropathy (PN) in obese non-diabetic leptin-deficient ob/ob mice. Neurol Res 2019; 41:341-353. [DOI: 10.1080/01616412.2018.1564191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Joanna Kosacka
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Katrin Woidt
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Klaus V. Toyka
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | - Sabine Paeschke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Integrated Research and Treatment Center (IFB) Adiposity Disease, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics (ILM), University of Leipzig, Leipzig, German
| | | | - Petra Baum
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Marcin Nowicki
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
39
|
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: 2.5] [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
|
40
|
Hubler MJ, Erikson KM, Kennedy AJ, Hasty AH. MFe hi adipose tissue macrophages compensate for tissue iron perturbations in mice. Am J Physiol Cell Physiol 2018; 315:C319-C329. [PMID: 29768045 DOI: 10.1152/ajpcell.00103.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Resident adipose tissue macrophages (ATMs) play multiple roles to maintain tissue homeostasis, such as removing excess free fatty acids and regulation of the extracellular matrix. The phagocytic nature and oxidative resiliency of macrophages not only allows them to function as innate immune cells but also to respond to specific tissue needs, such as iron homeostasis. MFehi ATMs are a subtype of resident ATMs that we recently identified to have twice the intracellular iron content as other ATMs and elevated expression of iron-handling genes. Although studies have demonstrated that iron homeostasis is important for adipocyte health, little is known about how MFehi ATMs may respond to and influence adipose tissue iron availability. Two methodologies were used to address this question: dietary iron supplementation and intraperitoneal iron injection. Upon exposure to high dietary iron, MFehi ATMs accumulated excess iron, whereas the iron content of MFelo ATMs and adipocytes remained unchanged. In this model of chronic iron excess, MFehi ATMs exhibited increased expression of genes involved in iron storage. In the injection model, MFehi ATMs incorporated high levels of iron, and adipocytes were spared iron overload. This acute model of iron overload was associated with increased numbers of MFehi ATMs; 17% could be attributed to monocyte recruitment and 83% to MFelo ATM incorporation into the MFehi pool. The MFehi ATM population maintained its low inflammatory profile and iron-cycling expression profile. These studies expand the field's understanding of ATMs and confirm that they can respond as a tissue iron sink in models of iron overload.
Collapse
Affiliation(s)
- Merla J Hubler
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University , Nashville, Tennessee
| | - Keith M Erikson
- Department of Nutrition, University of North Carolina at Greensboro , Greensboro, North Carolina
| | - Arion J Kennedy
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University , Nashville, Tennessee
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University , Nashville, Tennessee.,VA Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
41
|
Rodríguez-Pérez C, Vrhovnik P, González-Alzaga B, Fernández MF, Martin-Olmedo P, Olea N, Fiket Ž, Kniewald G, Arrebola JP. Socio-demographic, lifestyle, and dietary determinants of essential and possibly-essential trace element levels in adipose tissue from an adult cohort. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 236:878-888. [PMID: 29021094 DOI: 10.1016/j.envpol.2017.09.093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 06/07/2023]
Abstract
There is increasing evidence linking levels of trace elements (TEs) in adipose tissue with certain chronic conditions (e.g., diabetes or obesity). The objectives of this study were to assess concentrations of a selection of nine essential and possibly-essential TEs in adipose tissue samples from an adult cohort and to explore their socio-demographic, dietary, and lifestyle determinants. Adipose tissue samples were intraoperatively collected from 226 volunteers recruited in two public hospitals from Granada province. Trace elements (Co, Cr, Cu, Fe, Mn, Mo, Se, V, and Zn) were analyzed in adipose tissue by high-resolution inductively coupled plasma mass spectrometry (HR-ICP-MS). Data were collected on socio-demographic characteristics, lifestyle, diet, and health status by face-to-face interview. Predictors of TE concentrations were assessed by using multivariable linear and logistic regression. All TEs were detected in all samples with the exception of Se (53.50%). Iron, zinc, and copper showed the highest concentrations (42.60 mg/kg, 9.80 mg/kg, and 0.68 mg/kg, respectively). Diet was the main predictor of Cr, Fe, Mo, and Se concentrations. Body mass index was negatively associated with all TEs (β coefficients = -0.018 to -0.593, p = 0.001-0.090) except for Mn and V. Age showed a borderline-significant positive correlation with Cu (β = 0.004, p = 0.089). Residence in a rural or semi-rural area was associated with increased Co, Cr, Fe, Mo, Mn, V and Zn concentrations and with β coefficients ranging from 0.196 to 0.544 (p < 0.05). Furthermore, individuals with higher educational level showed increased Cr, Co, Fe and V concentrations (β coefficients = 0.276-0.368, p = 0.022-0.071). This is the first report on the distribution of these TEs in adipose tissue and on their determinants in a human cohort and might serve as an initial step in the elucidation of their clinical relevance.
Collapse
Affiliation(s)
| | - Petra Vrhovnik
- Slovenian National Building and Civil Engineering Institute (ZAG), Ljubljana, Slovenia
| | - Beatriz González-Alzaga
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Hospitales Universitarios de Granada, Spain; Andalusian School of Public Health (EASP), Granada, Spain
| | - Mariana F Fernández
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Hospitales Universitarios de Granada, Spain; University of Granada, Centro de Investigación Biomédica, Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Piedad Martin-Olmedo
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Hospitales Universitarios de Granada, Spain; Andalusian School of Public Health (EASP), Granada, Spain
| | - Nicolás Olea
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Hospitales Universitarios de Granada, Spain; University of Granada, Centro de Investigación Biomédica, Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Željka Fiket
- Ruđer Bošković, Division for Marine and Environmental Research, Zagreb, Croatia
| | - Goran Kniewald
- Ruđer Bošković, Division for Marine and Environmental Research, Zagreb, Croatia
| | - Juan P Arrebola
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Hospitales Universitarios de Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Oncology Unit, Virgen de las Nieves University Hospital, Granada, Spain.
| |
Collapse
|
42
|
Aguirre LG, Urrunaga-Pastor D, Moncada-Mapelli E, Guarnizo-Poma M, Lazaro-Alcantara H, Benites-Zapata VA. High serum ferritin levels are associated with insulin resistance but not with impaired glucose tolerance in a healthy people population. Diabetes Metab Syndr 2017; 11 Suppl 2:S983-S988. [PMID: 28755842 DOI: 10.1016/j.dsx.2017.07.026] [Citation(s) in RCA: 7] [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: 07/02/2017] [Accepted: 07/16/2017] [Indexed: 12/29/2022]
Abstract
AIM To assess the association between elevated serum ferritin levels and the presence of insulin resistance (IR) or impaired glucose tolerance (IGT) in a population of individuals with no endocrine or metabolic disorders background. METHODS Analytical cross-sectional study, carried out in adults of both sexes with no medical history of type 2 diabetes mellitus (T2DM) or other metabolic or endocrine disorder, who attended the outpatient service of a private clinic in Lima-Peru during 2012-2014 period. Impaired serum ferritin levels were defined as serum ferritin values >300μg/L in men and >200μg/L in women. IR was defined as a Homeostasis Model Assessment (HOMA-IR) value ≥3.8 and IGT was defined as an oral glucose tolerance test (OGTT) value between 126mg/dL and 199mg/dL. The reported association measure was the prevalence ratio (PR) with their respective 95% confidence intervals (95% CI). RESULTS We analyzed 213 participants, the average age was 35.8±11.1years and 35.7% were males. The prevalence of impaired serum ferritin levels, IR and IGT in the population was 12.7%, 33.3% and 9.9% respectively. In the adjusted Poisson regression models, the prevalence of IR was higher among the group with impaired serum ferritin levels (PR=1.74; 95%CI:1.18-2.56); however, we found no association between impaired serum ferritin levels and IGT (PR=1.42; 95%CI:0.47-4.30). CONCLUSIONS Impaired levels of serum ferritin are associated with IR, nevertheless, not with IGT in a metabolically healthy population. Serum ferritin could be considered as an early marker of IR prior to the onset of glycaemia disorders.
Collapse
Affiliation(s)
- Luis G Aguirre
- Sociedad Científica de Estudiantes de Medicina de la Universidad de San Martín de Porres, Universidad de San Martín de Porres, Lima, Peru
| | - Diego Urrunaga-Pastor
- Sociedad Científica de Estudiantes de Medicina de la Universidad de San Martín de Porres, Universidad de San Martín de Porres, Lima, Peru
| | - Enrique Moncada-Mapelli
- Sociedad Científica de Estudiantes de Medicina de la Universidad de San Martín de Porres, Universidad de San Martín de Porres, Lima, Peru
| | | | | | - Vicente A Benites-Zapata
- Centre for Public Health Research, Research Institute, Faculty of Medicine, Universidad de San Martín de Porres, Lima, Peru.
| |
Collapse
|
43
|
Abstract
Interactions between macrophages and adipocytes influence both metabolism and inflammation. Obesity-induced changes to macrophages and adipocytes lead to chronic inflammation and insulin resistance. This paper reviews the various functions of macrophages in lean and obese adipose tissue and how obesity alters adipose tissue macrophage phenotypes. Metabolic disease and insulin resistance shift the balance between numerous pro- and anti-inflammatory regulators of macrophages and create a feed-forward loop of increasing inflammatory macrophage activation and worsening adipocyte dysfunction. This ultimately leads to adipose tissue fibrosis and diabetes. The molecular mechanisms underlying these processes have therapeutic implications for obesity, metabolic syndrome, and diabetes.
Collapse
Affiliation(s)
- Dylan Thomas
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston Medical Center, 88 East Newton Street, H-3600, Boston, MA 02118.
| | - Caroline Apovian
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston Medical Center, 88 East Newton Street, Robinson 4400, Boston, MA 02118.
| |
Collapse
|
44
|
Chakraborty S, Bhattacharyya R, Banerjee D. Infections: A Possible Risk Factor for Type 2 Diabetes. Adv Clin Chem 2017; 80:227-251. [PMID: 28431641 DOI: 10.1016/bs.acc.2016.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Diabetes mellitus is one of the biggest challenges to human health globally, with an estimated 95% of the global diabetic population having type 2 diabetes. Classical causes for type 2 diabetes, such as genetics and obesity, do not account for the high incidence of the disease. Recent data suggest that infections may precipitate insulin resistance via multiple mechanisms, such as the proinflammatory cytokine response, the acute-phase response, and the alteration of the nutrient status. Even pathogen products, such as lipopolysaccharide and peptidoglycans, can be diabetogenic. Therefore, we argue that infections that are known to contribute to insulin resistance should be considered as risk factors for type 2 diabetes.
Collapse
Affiliation(s)
- Surajit Chakraborty
- Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Dibyajyoti Banerjee
- Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
45
|
Wang CM, Chen YH, Lee YC, Chang JS. Endoplasmic reticulum stress contributes to ferritin molecules-mediated macrophage migration via P-selectin glycoprotein ligand-1. Mol Nutr Food Res 2016; 61. [DOI: 10.1002/mnfr.201600458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 01/11/2023]
Affiliation(s)
- Chi-Mei Wang
- Department of Nutrition; MacKay Memorial Hospital; Taiwan
| | - Yue-Hwa Chen
- School of Nutrition and Health Sciences; College of Nutrition; Taipei Medical University; Taipei Taiwan
- Nutrition Research Center; Taipei Medical University Hospital; Taipei Taiwan
| | - Yu-Chieh Lee
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Jung-Su Chang
- School of Nutrition and Health Sciences; College of Nutrition; Taipei Medical University; Taipei Taiwan
- Nutrition Research Center; Taipei Medical University Hospital; Taipei Taiwan
| |
Collapse
|
46
|
Kimura Y, Yasuda K, Kurotani K, Akter S, Kashino I, Hayabuchi H, Sato M, Mizoue T. Circulating ferritin concentrations are differentially associated with serum adipokine concentrations in Japanese men and premenopausal women. Eur J Nutr 2016; 56:2497-2505. [PMID: 27484685 DOI: 10.1007/s00394-016-1285-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 07/26/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Increased iron storage, as measured by circulating ferritin, has been linked to an increased risk of various diseases including diabetes. We examined the association of circulating ferritin with serum adiponectin, leptin, resistin, plasminogen activator inhibitor-1 (PAI-1), and visfatin levels. METHODS We conducted a cross-sectional study among 429 Japanese employees (284 men and 145 premenopausal women, mean age: 42.5 ± 10.5 years). Serum adipokines were measured using Luminex suspension bead-based multiplexed array, and serum ferritin was determined using a chemiluminescence immunoassay. Multivariable regression analysis was performed to calculate mean concentrations of adipokine according to the tertile of ferritin concentrations with adjustment for potential confounders. RESULTS Leptin and visfatin concentrations increased with increasing ferritin concentrations in men after multivariable adjustment of physical activity, smoking, alcohol use, and body mass index (P for trend = 0.02 and 0.01 for leptin and visfatin, respectively). Serum ferritin concentrations were inversely and significantly associated with adiponectin in women (P for trend = 0.01). Resistin and PAI-1 were not appreciably associated with ferritin concentration. CONCLUSIONS Increased iron storage may be associated with higher circulating concentrations of leptin and visfatin in men and with lower concentrations of adiponectin in women.
Collapse
Affiliation(s)
- Yasumi Kimura
- Department of Nutritional Sciences, Faculty of Nutritional Sciences, Nakamura Gakuen University, 5-7-1 Befu, Jounan-Ku, Fukuoka, 814-0104, Japan.
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kayo Kurotani
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shamima Akter
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ikuko Kashino
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hitomi Hayabuchi
- Graduate School of Nutrition and Health Science, Fukuoka Women's University, Fukuoka, Japan
| | - Masao Sato
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
47
|
Gotardo ÉMF, Caria CREP, de Oliveira CC, Rocha T, Ribeiro ML, Gambero A. Effects of iron supplementation in mice with hypoferremia induced by obesity. Exp Biol Med (Maywood) 2016; 241:2049-2055. [PMID: 27439539 DOI: 10.1177/1535370216660398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Iron is an important micronutrient, but it can also act as a dangerous element by interfering with glucose homeostasis and inflammation, two features that are already disturbed in obese subjects. In this work, we study the effects of systemic iron supplementation on metabolic and inflammatory responses in mice with hypoferremia induced by obesity to better characterize whether iron worsens the parameters that are already altered after 24 weeks of a high-fat diet (HFD). Mice were maintained on a control diet or a HFD for 24 weeks and received iron-III polymaltose (50 mg/kg/every 2 days) during the last two weeks. Glucose homeostasis (basal glucose and insulin test tolerance) and systemic and visceral adipose tissue (VAT) inflammation were assessed. Iron levels were measured in serum. The Prussian blue reaction was used in isolated macrophages to detect iron deposition. Iron supplementation resulted in an increased number of VAT macrophages that were positive for Prussian blue staining as well as increased serum iron levels. Systemic hepcidin, leptin, resistin, and monocyte chemoattractant protein-1 (MCP-1) levels were not altered by iron supplementation. Local adipose tissue inflammation was also not made worse by iron supplementation because the levels of hepcidin, MCP-1, leptin, and interleukin (IL)-6 were not altered. In contrast, iron supplementation resulted in an increased production of IL-10 by adipose tissue and VAT macrophages. Leukocytosis and VAT plasminogen activator inhibitor-1 (PAI-1) level were reduced, but insulin resistance was not altered after iron supplementation. In conclusion, systemic iron supplementation in mice with hypoferremia induced by obesity did not worsen inflammatory marker or adipose tissue inflammation or the metabolic status established by obesity. Iron deposition was observed in adipose tissue, mainly in macrophages, suggesting that these cells have mechanisms that promote iron incorporation without increasing the production of inflammatory mediators.
Collapse
Affiliation(s)
- Érica Martins Ferreira Gotardo
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista 12916-900, SP, Brazil
| | - Cintia Rabelo E Paiva Caria
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista 12916-900, SP, Brazil
| | - Caroline Candida de Oliveira
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista 12916-900, SP, Brazil
| | - Thalita Rocha
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista 12916-900, SP, Brazil
| | - Marcelo Lima Ribeiro
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista 12916-900, SP, Brazil
| | - Alessandra Gambero
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista 12916-900, SP, Brazil
| |
Collapse
|
48
|
Sharma NK, Sajuthi SP, Chou JW, Calles-Escandon J, Demons J, Rogers S, Ma L, Palmer ND, McWilliams DR, Beal J, Comeau ME, Cherry K, Hawkins GA, Menon L, Kouba E, Davis D, Burris M, Byerly SJ, Easter L, Bowden DW, Freedman BI, Langefeld CD, Das SK. Tissue-Specific and Genetic Regulation of Insulin Sensitivity-Associated Transcripts in African Americans. J Clin Endocrinol Metab 2016; 101:1455-68. [PMID: 26789776 PMCID: PMC4880154 DOI: 10.1210/jc.2015-3336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Integrative multiomics analyses of adipose and muscle tissue transcripts, S, and genotypes revealed novel genetic regulatory mechanisms of insulin resistance in African Americans.
Collapse
Affiliation(s)
- Neeraj K Sharma
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Satria P Sajuthi
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Jeff W Chou
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Jorge Calles-Escandon
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Jamehl Demons
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Samantha Rogers
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Lijun Ma
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Nicholette D Palmer
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - David R McWilliams
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - John Beal
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Mary E Comeau
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Kristina Cherry
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Gregory A Hawkins
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Lata Menon
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Ethel Kouba
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Donna Davis
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Marcie Burris
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Sara J Byerly
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Linda Easter
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Donald W Bowden
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Barry I Freedman
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Carl D Langefeld
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Swapan K Das
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| |
Collapse
|
49
|
Iron Overload Coordinately Promotes Ferritin Expression and Fat Accumulation in Caenorhabditis elegans. Genetics 2016; 203:241-53. [PMID: 27017620 DOI: 10.1534/genetics.116.186742] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/20/2016] [Indexed: 01/22/2023] Open
Abstract
The trace element iron is crucial for living organisms, since it plays essential roles in numerous cellular functions. Systemic iron overload and the elevated level of ferritin, a ubiquitous intracellular protein that stores and releases iron to maintain the iron homeostasis in cells, has long been epidemiologically associated with obesity and obesity-related diseases. However, the underlying mechanisms of this association remain unclear. Here, using Caenorhabditis elegans, we show that iron overload induces the expression of sgk-1, encoding the serum and glucocorticoid-inducible kinase, to promote the level of ferritin and fat accumulation. Mutation of cyp-23A1, encoding a homolog of human cytochrome P450 CYP7B1 that is related to neonatal hemochromatosis, further enhances the elevated expression of ftn-1, sgk-1, and fat accumulation. sgk-1 positively regulates the expression of acs-20 and vit-2, genes encoding homologs of the mammalian FATP1/4 fatty acid transport proteins and yolk lipoproteins, respectively, to facilitate lipid uptake and translocation for storage under iron overload. This study reveals a completely novel pathway in which sgk-1 plays a central role to synergistically regulate iron and lipid homeostasis, offering not only experimental evidence supporting a previously unverified link between iron and obesity, but also novel insights into the pathogenesis of iron and obesity-related human metabolic diseases.
Collapse
|
50
|
Recalcati S, Gammella E, Cairo G. New perspectives on the molecular basis of the interaction between oxygen homeostasis and iron metabolism. HYPOXIA 2015; 3:93-103. [PMID: 27774486 PMCID: PMC5045093 DOI: 10.2147/hp.s83537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxygen and iron are two elements closely related from a (bio)chemical point of view. Moreover, they share the characteristic of being indispensable for life, while also being potentially toxic. Therefore, their level is strictly monitored, and sophisticated pathways have evolved to face variations in either element. In addition, the expression of proteins involved in iron and oxygen metabolism is mainly controlled by a complex interplay of proteins that sense both iron levels and oxygen availability (ie, prolyl hydroxylases, hypoxia inducible factors, and iron regulatory proteins), and in turn activate feedback mechanisms to re-establish homeostasis. In this review, we describe how cells and organisms utilize these intricate networks to regulate responses to changes in oxygen and iron levels. We also explore the role of these pathways in some pathophysiological settings.
Collapse
Affiliation(s)
- Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|