1
|
Sharma S, Kumar S, Tomar MS, Chauhan D, Kulkarni C, Rajput S, Sadhukhan S, Porwal K, Guha R, Shrivastava A, Gayen JR, Kumar N, Chattopadhyay N. Multiscale effects of the calcimimetic drug, etelcalcetide on bone health of rats with secondary hyperparathyroidism induced by chronic kidney disease. Bone 2024; 185:117126. [PMID: 38777312 DOI: 10.1016/j.bone.2024.117126] [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: 02/14/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Chronic kidney disease-induced secondary hyperparathyroidism (CKD-SHPT) heightens fracture risk through impaired mineral homeostasis and elevated levels of uremic toxins (UTs), which in turn enhance bone remodeling. Etelcalcetide (Etel), a calcium-sensing receptor (CaSR) agonist, suppresses parathyroid hormone (PTH) in hyperparathyroidism to reduce excessive bone resorption, leading to increased bone mass. However, Etel's effect on bone quality, chemical composition, and strength is not well understood. To address these gaps, we established a CKD-SHPT rat model and administered Etel at a human-equivalent dose concurrently with disease induction. The effects on bone and mineral homeostasis were compared with a CKD-SHPT (vehicle-treated group) and a control group (rats without SHPT). Compared with vehicle-treated CKD-SHPT rats, Etel treatment improved renal function, reduced circulating UT levels, improved mineral homeostasis parameters, decreased PTH levels, and prevented mineralization defects. The upregulation of mineralization-promoting genes by Etel in CKD-SHPT rats might explain its ability to prevent mineralization defects. Etel preserved both trabecular and cortical bones with attendant suppression of osteoclast function, besides increasing mineralization. Etel maintained the number of viable osteocytes to the control level, which could also contribute to its beneficial effects on bone. CKD-SHPT rats displayed increased carbonate substitution of matrix and mineral, decreased crystallinity, mineral-to-matrix ratio, and collagen maturity, and these changes were mitigated by Etel. Further, Etel treatment prevented CKD-SHPT-induced deterioration in bone strength and mechanical behavior. Based on these findings, we conclude that in CKD-SHPT rats, Etel has multiscale beneficial effects on bone that involve remodeling suppression, mineralization gene upregulation, and preservation of osteocytes.
Collapse
Affiliation(s)
- Shivani Sharma
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Saroj Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow 226003, India
| | - Divya Chauhan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India
| | - Chirag Kulkarni
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Swati Rajput
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sreyanko Sadhukhan
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Konica Porwal
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India
| | - Rajdeep Guha
- Division of Laboratory Animal Facility, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow 226003, India
| | - Jiaur R Gayen
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India
| | - Navin Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Samaha MM, Nour OA, Sewilam HM, El-Kashef DH. Diacerein mitigates adenine-induced chronic kidney disease in rats: Focus on TLR4/MYD88/TRAF6/NF-κB pathway. Life Sci 2023; 331:122080. [PMID: 37690574 DOI: 10.1016/j.lfs.2023.122080] [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: 06/18/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Chronic kidney disease (CKD) is a serious problem which negatively affects human health. AIMS The purpose of this investigation was to explore the possible beneficial impacts of diacerein on adenine-induced CKD in rats. MAIN METHODS 32 male Sprague Dawley rats were allocated into 4 groups; normal, diseased (200 mg/kg adenine, orally) and diacerein (25 and 50 mg/kg, orally). KEY FINDINGS Adenine produced marked reduction in rats' body weights and a substantial increase in kidney/body weight index. Additionally, adenine significantly increased serum creatinine and BUN levels besides proteinuria levels, and also reduced creatinine clearance. Adenine induced oxidative stress as evidenced by increased MDA content and diminished GSH concentration in renal tissues. These biochemical measurements were confirmed by the morphological and histopathological results. Moreover, adenine revealed substantial elevation in renal level and expression of MYD88, TRAF6 and TNF-α, and renal level of IL-1β in addition to increased expression of TLR4, NF-κB p65 and p-NF-κB p65 while reduced the expression of IκB-α. Diacerein in a dose-dependent manner effectively ameliorated adenine-induced alterations. SIGNIFICANCE Diacerein could be used as a therapeutic agent to attenuate CKD after further clinical studies.
Collapse
Affiliation(s)
- Mahmoud M Samaha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Omnia A Nour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Haitham M Sewilam
- Department of Histology, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
3
|
Grander M, Hoffmann A, Seifert M, Demetz E, Grubwieser P, Pfeifhofer-Obermair C, Haschka D, Weiss G. DMT1 Protects Macrophages from Salmonella Infection by Controlling Cellular Iron Turnover and Lipocalin 2 Expression. Int J Mol Sci 2022; 23:ijms23126789. [PMID: 35743233 PMCID: PMC9223531 DOI: 10.3390/ijms23126789] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Macrophages are at the center of innate pathogen control and iron recycling. Divalent metal transporter 1 (DMT1) is essential for the uptake of non-transferrin-bound iron (NTBI) into macrophages and for the transfer of transferrin-bound iron from the endosome to the cytoplasm. As the control of cellular iron trafficking is central for the control of infection with siderophilic pathogens such as Salmonella Typhimurium, a Gram-negative bacterium residing within the phagosome of macrophages, we examined the potential role of DMT1 for infection control. Bone marrow derived macrophages lacking DMT1 (DMT1fl/flLysMCre(+)) present with reduced NTBI uptake and reduced levels of the iron storage protein ferritin, the iron exporter ferroportin and, surprisingly, of the iron uptake protein transferrin receptor. Further, DMT1-deficient macrophages have an impaired control of Salmonella Typhimurium infection, paralleled by reduced levels of the peptide lipocalin-2 (LCN2). LCN2 exerts anti-bacterial activity upon binding of microbial siderophores but also facilitates systemic and cellular hypoferremia. Remarkably, nifedipine, a pharmacological DMT1 activator, stimulates LCN2 expression in RAW264.7 macrophages, confirming its DMT1-dependent regulation. In addition, the absence of DMT1 increases the availability of iron for Salmonella upon infection and leads to increased bacterial proliferation and persistence within macrophages. Accordingly, mice harboring a macrophage-selective DMT1 disruption demonstrate reduced survival following Salmonella infection. This study highlights the importance of DMT1 in nutritional immunity and the significance of iron delivery for the control of infection with siderophilic bacteria.
Collapse
Affiliation(s)
- Manuel Grander
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Alexander Hoffmann
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
| | - Philipp Grubwieser
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Correspondence: (D.H.); (G.W.)
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.G.); (A.H.); (M.S.); (E.D.); (P.G.); (C.P.-O.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (D.H.); (G.W.)
| |
Collapse
|
4
|
Zhang Y, Mou Y, Zhang J, Suo C, Zhou H, Gu M, Wang Z, Tan R. Therapeutic Implications of Ferroptosis in Renal Fibrosis. Front Mol Biosci 2022; 9:890766. [PMID: 35655759 PMCID: PMC9152458 DOI: 10.3389/fmolb.2022.890766] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/12/2022] [Indexed: 12/13/2022] Open
Abstract
Renal fibrosis is a common feature of chronic kidney disease (CKD), and can lead to the destruction of normal renal structure and loss of kidney function. Little progress has been made in reversing fibrosis in recent years. Ferroptosis is more immunogenic than apoptosis due to the release and activation of damage-related molecular patterns (DAMPs) signals. In this paper, the relationship between renal fibrosis and ferroptosis was reviewed from the perspective of iron metabolism and lipid peroxidation, and some pharmaceuticals or chemicals associated with both ferroptosis and renal fibrosis were summarized. Other programmed cell death and ferroptosis in renal fibrosis were also firstly reviewed for comparison and further investigation.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhua Mou
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jianjian Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanjian Suo
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai Zhou
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Gu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Ruoyun Tan,
| |
Collapse
|
5
|
Comparison of Stromal Vascular Fraction and Adipose-Derived Stem Cells for Protection of Renal Function in a Rodent Model of Ischemic Acute Kidney Injury. Stem Cells Int 2022; 2022:1379680. [PMID: 35578662 PMCID: PMC9107055 DOI: 10.1155/2022/1379680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Aims Few studies have compared the use of different cell types derived from adipose tissue or the optimal route for efficient and safe cell delivery in ischemic acute kidney injury (AKI). We compared the abilities of stromal vascular fraction (SVF) and adipose-derived stem cells (ADSC), injected via three different routes, to protect renal function in a rodent model of ischemic AKI. Methods Ninety male Sprague-Dawley rats were randomly divided into 9 groups: sham, nephrectomy control, AKI control, transaortic renal arterial SVF injection, renal parenchymal SVF injection, tail venous SVF injection, transaortic renal arterial ADSC injection, renal parenchymal ADSC injection, and tail venous ADSC injection groups. Their renal function was assessed 4 days before and 1, 2, 3, 4, 7, and 14 days after surgical procedures to induce ischemic AKI. The histomorphometric studies were performed 14 days after surgical procedures. Results Renal parenchymal injection of SVF notably reduced the level of serum blood urea nitrogen and creatinine elevation compared to the AKI control group. Renal parenchymal injection of SVF notably reduced the level of creatinine clearance decrease. In addition, collagen content was lower in the renal parenchymal SVF injection group, and fibrosis was reduced. Apoptosis was reduced in the renal parenchymal SVF injection group, and proliferation was increased. The expression levels of antioxidative markers such as glutathione reductase and peroxidase were higher in the renal parenchymal SVF injection group. Conclusions Our findings suggest that renal function is protected from ischemic AKI through renal parenchymal injection of SVF, which has enhanced antifibrotic, antiapoptotic, and antioxidative effects.
Collapse
|
6
|
Integrated gut microbiota and fecal metabolomics reveal the renoprotective effect of Rehmanniae Radix Preparata and Corni Fructus on adenine-induced CKD rats. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1174:122728. [PMID: 33975272 DOI: 10.1016/j.jchromb.2021.122728] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/18/2021] [Accepted: 04/14/2021] [Indexed: 12/27/2022]
Abstract
Rehmanniae Radix Preparata (RR) and Corni Fructus (CF), well-known traditional Chinese medicines (TCMs), were generally used together in the clinical practices to treat chronic kidney disease (CKD) with synergistic effects for thousands of years, but their combination mechanism remains largely unknown so far. Recent evidences have implicated intestinal flora as potential targets for the therapy of CKD. In this study, the CKD rat model was induced by adenine. The levels of proteinuria, serum creatine (SCr), blood urea nitrogen (BUN) and creatinine clearance (Ccr) were used to assess the cooperation effect of RR and CF. Furthermore, high-throughput 16S ribosomal RNA (rRNA) gene sequencing combined with fecal metabonomics based on UPLC-Q-TOF-MS/MS were applied to explore the variations of intestinal flora and their metabolic profiles. 16S rRNA gene sequencing data indicated that CKD rats treated with RR, CF and RC showed the differences in the composition of gut microbiota. The abundance of beneficial bacteria including Ruminococcaceae UCG-014, Ruminococcus 1, Prevotellaceae_NK3B31_group, Lachnospiraceae NK4A136 group and Lachnospiraceae UCG-001 were elevated in various degrees, while the opportunistic pathogen such as Desulfovibrio was markedly decreased after the treatment. Moreover, fecal metabolite profiles revealed 15 different metabolites associated with CKD. These metabolites were mainly involved in the related metabolic pathways such as amino acid metabolism, bile acids metabolism and glycerophospholipid metabolism. The results implied that gut flora and their metabolites might play a vital role in the progress of CKD, which provided a potential target for the development of novel drugs for the therapy of CKD.
Collapse
|
7
|
Nemmar A, Beegam S, Yuvaraju P, Yasin J, Ali BH, Adeghate E. Nose-Only Water-Pipe Smoke Exposure in Mice Elicits Renal Histopathological Alterations, Inflammation, Oxidative Stress, DNA Damage, and Apoptosis. Front Physiol 2020; 11:46. [PMID: 32116758 PMCID: PMC7026484 DOI: 10.3389/fphys.2020.00046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of water-pipe tobacco smoking is increasing worldwide, and is relatively high among youth and young adults. Exposure to water-pipe smoke (WPS) has been reported to affect various systems including the respiratory, cardiovascular and reproductive systems. However, the impact of WPS exposure on the kidney has received only scant attention. Here, we assessed the effect of nose-only WPS exposure for one or four consecutive weeks on renal histology, inflammation, oxidative stress, DNA damage, and apoptosis. The duration of the session was 30 min/day and 5 days/week. Control mice were exposed to air. Light and electron microcopy analysis revealed that the WPS exposure (especially at 4-week time point) caused degeneration of the endothelial cells of the glomerular capillaries and vacuolar degenerations of the proximal convoluted tubules. WPS exposure also significantly decreased the creatinine clearance, and significantly increased proteinuria and urinary kidney injury molecule-1 (KIM-1) concentration. Kidney lipid peroxidation, reactive oxygen species, and oxidized glutathione were significantly increased. WPS exposure also affected the concentration of reduced glutathione and the activity of catalase. Likewise, renal concentrations of interleukin (IL)-6, IL-1β and KIM-1 were augmented by WPS exposure. Moreover, WPS caused DNA damage as evaluated by comet assay, and increased the expression of cleaved caspase-3 and cytochrome C in the kidney. We conclude that exposure of mice to WPS caused renal histopathological alterations, inflammation, oxidative stress, DNA damage, and apoptosis. If the latter findings could be substantiated by controlled human studies, it would be an additional cause for disquiet about an established public health concern.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Priya Yuvaraju
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Javed Yasin
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
8
|
Abdelrahman AM, Al Salam S, Al Suleimani Y, Ashique M, Manoj P, Ali BH. Effect of levosimendan, an inodilator, on streptozotocin-induced diabetic nephropathy in rats. Eur J Pharmacol 2020; 873:172960. [PMID: 32001219 DOI: 10.1016/j.ejphar.2020.172960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/06/2020] [Accepted: 01/24/2020] [Indexed: 01/26/2023]
Abstract
This study examined the effect of levosimendan on streptozotocin-induced early diabetic nephropathy. Rats were distributed into four groups and treated for six weeks. The first and third group received either vehicle or levosimendan (1 mg/kg/day) for the last three weeks, respectively. The second and fourth groups were rendered diabetic by a single intraperitoneal injection of streptozotocin (60 mg/kg) and were treated as the first and third groups, respectively. In the untreated diabetic group, there was a significant decrease in body weight, polyuria and hyperglycemia as well as, increased urinary albumin/creatinine ratio (UACR) and N-acetyl-β-D-glucosaminidase (NAG)/creatinine ratio (UNCR) with no change in creatinine clearance. In addition, diabetes was associated with increased oxidative stress as evidenced by reduced plasma total antioxidant capacity (TAC) and catalase activity and increased plasma malondialdhyde (MDA) and the inflammatory marker, tumor necrosis factor-alpha, (TNF-α). Kidneys from streptozotocin-treated rats showed focal clear renal tubular cells affecting proximal convoluted tubules and mild interstitial fibrosis at the cortico-medullary junction. Levosimendan significantly attenuated the streptozotocin-induced physiological and biochemical changes and there was less clear renal tubular cells. This study shows that levosimendan ameliorated some of the changes seen in streptozotocin-induced early diabetic nephropathy in rats. This could be partly due to its antioxidative and anti-inflammatory effects.
Collapse
Affiliation(s)
- Aly M Abdelrahman
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod, Oman.
| | - Suhail Al Salam
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Yousuf Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod, Oman
| | - Mohamed Ashique
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod, Oman
| | - Priyadarsini Manoj
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod, Oman
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod, Oman
| |
Collapse
|
9
|
Wang X, Zhang M, Flores SRL, Woloshun RR, Yang C, Yin L, Xiang P, Xu X, Garrick MD, Vidyasagar S, Merlin D, Collins JF. Oral Gavage of Ginger Nanoparticle-Derived Lipid Vectors Carrying Dmt1 siRNA Blunts Iron Loading in Murine Hereditary Hemochromatosis. Mol Ther 2019; 27:493-506. [PMID: 30713087 PMCID: PMC6401192 DOI: 10.1016/j.ymthe.2019.01.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles (NPs) have been utilized to deliver drugs to the intestinal epithelium in vivo. Moreover, NPs derived from edible plants are less toxic than synthetic NPs. Here, we utilized ginger NP-derived lipid vectors (GDLVs) in a proof-of-concept investigation to test the hypothesis that inhibiting expression of divalent metal-ion transporter 1 (Dmt1) would attenuate iron loading in a mouse model of hereditary hemochromatosis (HH). Initial experiments using duodenal epithelial organ cultures from intestine-specific Dmt1 knockout (KO) (Dmt1int/int) mice in the Ussing chamber established that Dmt1 is the only active iron importer during iron-deficiency anemia. Further, when Dmt1int/int mice were crossed with mice lacking the iron-regulatory hormone, hepcidin (Hepc-/-), iron loading was abolished. Hence, intestinal Dmt1 is required for the excessive iron absorption that typifies HH. Additional experiments established a protocol to produce GDLVs carrying functional Dmt1 small interfering RNAs (siRNAs) and to target these gene delivery vehicles to the duodenal epithelium in vivo (by incorporating folic acid [FA]). When FA-GDLVs carrying Dmt1 siRNA were administered to weanling Hepc-/- mice for 16 days, intestinal Dmt1 mRNA expression was attenuated and tissue iron accumulation was blunted. Oral delivery of functional siRNAs by FA-GDLVs is a suitable therapeutic approach to mitigate iron loading in murine HH.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Mingzhen Zhang
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Shireen R L Flores
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Regina R Woloshun
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Chunhua Yang
- Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Liangjie Yin
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - Ping Xiang
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA; State Key Lab of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Xiaodong Xu
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - Michael D Garrick
- Department of Biochemistry, State University of New York (SUNY), Buffalo, NY, USA
| | | | - Didier Merlin
- Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - James F Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
10
|
Vela D. Systemic and local hepcidin as emerging and important peptides in renal homeostasis and pathology. Biofactors 2019; 45:118-134. [PMID: 30461080 DOI: 10.1002/biof.1468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022]
Abstract
Recent data suggest that the importance of hepcidin goes beyond its classical role in controlling systemic iron metabolism. Local hepcidins are emerging as important peptides for organ homeostasis in the brain, heart, blood vessels, and in cancer as well. Similarly, accumulating data indicate that hepcidin does seem to be an important factor in renal homeostasis. This review encompasses present knowledge concerning the role of hepcidin in renoprotection and its use as a biomarker of kidney diseases. Understanding the role of hepcidin in kidneys is important due to its relevance for kidney physiology and its potential therapeutic application in kidney pathologies. © 2018 BioFactors, 45(2):118-134, 2019.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, Faculty of Medicine, University of Prishtina, Prishtina, Kosova
| |
Collapse
|
11
|
Abstract
Most cells in the body acquire iron via receptor-mediated endocytosis of transferrin, the circulating iron transport protein. When cellular iron levels are sufficient, the uptake of transferrin decreases to limit further iron assimilation and prevent excessive iron accumulation. In iron overload conditions, such as hereditary hemochromatosis and thalassemia major, unregulated iron entry into the plasma overwhelms the carrying capacity of transferrin, resulting in non-transferrin-bound iron (NTBI), a redox-active, potentially toxic form of iron. Plasma NTBI is rapidly cleared from the circulation primarily by the liver and other organs (e.g., pancreas, heart, and pituitary) where it contributes significantly to tissue iron overload and related pathology. While NTBI is usually not detectable in the plasma of healthy individuals, it does appear to be a normal constituent of brain interstitial fluid and therefore likely serves as an important source of iron for most cell types in the CNS. A growing body of literature indicates that NTBI uptake is mediated by non-transferrin-bound iron transporters such as ZIP14, L-type and T-type calcium channels, DMT1, ZIP8, and TRPC6. This review provides an overview of NTBI uptake by various tissues and cells and summarizes the evidence for and against the roles of individual transporters in this process.
Collapse
Affiliation(s)
- Mitchell D Knutson
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Abdelrahman AM, Suleimani YA, Za'abi MA, Ashique M, Manoj P, Hartmann C, Nemmar A, Schupp N, Ali BH. The renoprotective effect of the dipeptidyl peptidase-4 inhibitor sitagliptin on adenine-induced kidney disease in rats. Biomed Pharmacother 2019; 110:667-676. [DOI: 10.1016/j.biopha.2018.11.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/13/2018] [Accepted: 11/25/2018] [Indexed: 12/18/2022] Open
|
13
|
Tubular iron deposition and iron handling proteins in human healthy kidney and chronic kidney disease. Sci Rep 2018; 8:9353. [PMID: 29921869 PMCID: PMC6008459 DOI: 10.1038/s41598-018-27107-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/23/2018] [Indexed: 02/07/2023] Open
Abstract
Iron is suggested to play a detrimental role in the progression of chronic kidney disease (CKD). The kidney recycles iron back into the circulation. However, the localization of proteins relevant for physiological tubular iron handling and their potential role in CKD remain unclear. We examined associations between iron deposition, expression of iron handling proteins and tubular injury in kidney biopsies from CKD patients and healthy controls using immunohistochemistry. Iron was deposited in proximal (PT) and distal tubules (DT) in 33% of CKD biopsies, predominantly in pathologies with glomerular dysfunction, but absent in controls. In healthy kidney, PT contained proteins required for iron recycling including putative iron importers ZIP8, ZIP14, DMT1, iron storage proteins L- and H-ferritin and iron exporter ferroportin, while DT only contained ZIP8, ZIP14, and DMT1. In CKD, iron deposition associated with increased intensity of iron importers (ZIP14, ZIP8), storage proteins (L-, H-ferritin), and/or decreased ferroportin abundance. This demonstrates that tubular iron accumulation may result from increased iron uptake and/or inadequate iron export. Iron deposition associated with oxidative injury as indicated by heme oxygenase-1 abundance. In conclusion, iron deposition is relatively common in CKD, and may result from altered molecular iron handling and may contribute to renal injury.
Collapse
|
14
|
Abstract
Hypertension and chronic kidney disease (CKD) have a significant impact on global morbidity and mortality. The Low Birth Weight and Nephron Number Working Group has prepared a consensus document aimed to address the relatively neglected issue for the developmental programming of hypertension and CKD. It emerged from a workshop held on April 2, 2016, including eminent internationally recognized experts in the field of obstetrics, neonatology, and nephrology. Through multidisciplinary engagement, the goal of the workshop was to highlight the association between fetal and childhood development and an increased risk of adult diseases, focusing on hypertension and CKD, and to suggest possible practical solutions for the future. The recommendations for action of the consensus workshop are the results of combined clinical experience, shared research expertise, and a review of the literature. They highlight the need to act early to prevent CKD and other related noncommunicable diseases later in life by reducing low birth weight, small for gestational age, prematurity, and low nephron numbers at birth through coordinated interventions. Meeting the current unmet needs would help to define the most cost-effective strategies and to optimize interventions to limit or interrupt the developmental programming cycle of CKD later in life, especially in the poorest part of the world.
Collapse
|
15
|
Yu SS, Jiang LR, Ling Y, Qian ZM, Zhou YF, Li J, Ke Y. Nifedipine Increases Iron Content in WKPT-0293 Cl.2 Cells via Up-Regulating Iron Influx Proteins. Front Pharmacol 2017; 8:60. [PMID: 28243203 PMCID: PMC5303744 DOI: 10.3389/fphar.2017.00060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/30/2017] [Indexed: 12/25/2022] Open
Abstract
Nifedipine was reported to enhance urinary iron excretion in iron overloaded mice. However, it remains unknown how nifedipine stimulates urinary iron excretion in the kidney. We speculated that nifedipine might inhibit the TfR1/ DMT1 (transferrin receptor 1/divalent metal transporter1)-mediated iron uptake by proximal tubule cells in addition to blocking L-type Ca2+ channels, leading to an increase in iron in lumen-fluid and then urinary iron excretion. To test this hypothesis, we investigated the effects of nifedipine on iron content and expression of TfR1, DMT1 and ferroportin1 (Fpn1) in WKPT-0293 Cl.2 cells of the S1 segment of the proximal tubule in rats, using a graphite furnace atomic absorption spectrophotometer and Western blot analysis, respectively. We demonstrated for the first time that nifedipine significantly enhanced iron content as well as TfR1 and DMT1 expression and had no effect on Fpn1 levels in the cells. We also found that ferric ammonium citrate decreased TfR1 levels, increased Fpn1 expression and had no effect on DMT1 content, while co-treatment with nifedipine and FAC increase TfR1 and DMT1 expression and also had no effect on Fpn1 levels. These findings suggest that the nifedipine-induced increase in cell iron may mainly be due to the corresponding increase in TfR1 and DMT1 expression and also imply that the effects of nifedipine on iron transport in proximal tubule cells can not explain the increase in urinary iron excretion.
Collapse
Affiliation(s)
- Shuang-Shuang Yu
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Li-Rong Jiang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Yan Ling
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Juan Li
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong Hong Kong, Hong Kong
| |
Collapse
|
16
|
Tavella A, Stefani A, Zanardello C, Bettini A, Gauly M, Zanolari P. Dystrophic mineralization of the arterial fibrovascular tissue associated with a vitamin D hypervitaminosis in an 8-year-old female Alpaca ( Vicugna pacos). Ir Vet J 2016; 69:19. [PMID: 28018578 PMCID: PMC5160019 DOI: 10.1186/s13620-016-0078-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/01/2016] [Indexed: 02/01/2023] Open
Abstract
Background Prophylactic Vitamin D supplementation is a common practice in Alpaca breeding in many regions around the world. An overdosage can lead to dystrophic mineralization of soft tissues. In this paper we illustrate a suspected case of hypervitaminosis D in an 8-year-old female Alpaca. Case presentation In June 2015, the carcass of an 8-year-old female Alpaca (Vicugna pacos) was submitted to the diagnostic laboratory of the Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe) for necropsy. The animal had undergone a spontaneous abortion with uterine prolapse and delivery of the placenta, and had died shortly thereafter. Death occurred due to internal haemorrhage related to dystrophic mineralization of the left renal artery with subsequent rupture and damage of the renal hilum. During the necropsy, histopathological and serum biochemical analyses were performed. After laboratory analyses and the history of mineral and vitamin supplementation reported by the breeder, a hypervitaminosis D was suspected to be the most probable cause of the dystrophic mineralization observed in the left renal artery. Conclusions Most of the information regarding Llamas and Alpacas comes from the South American and Australian regions. It is therefore important to provide scientific information about these animals in other regions of the world in order to have a better and wider understanding of the nutritional and environmental conditions necessary for optimal breeding.
Collapse
Affiliation(s)
- Alexander Tavella
- Istituto Zooprofilattico Sperimentale delle Venezie, viale dell'Università 10, 35020 Legnaro, Italy
| | - Annalisa Stefani
- Istituto Zooprofilattico Sperimentale delle Venezie, viale dell'Università 10, 35020 Legnaro, Italy
| | - Claudia Zanardello
- Istituto Zooprofilattico Sperimentale delle Venezie, viale dell'Università 10, 35020 Legnaro, Italy
| | - Astrid Bettini
- Istituto Zooprofilattico Sperimentale delle Venezie, viale dell'Università 10, 35020 Legnaro, Italy
| | - Matthias Gauly
- Libera Università di Bolzano, Department of Animal Science, Faculty of Science and Technology, Piazza Università 5, 39100 Bolzano, Italy
| | - Patrik Zanolari
- Clinic for Ruminants, Vetsuisse-Faculty of the University of Bern, Bremgartenstrasse 109A, 3012 Bern, Switzerland
| |
Collapse
|
17
|
Thévenod F, Wolff NA. Iron transport in the kidney: implications for physiology and cadmium nephrotoxicity. Metallomics 2016; 8:17-42. [PMID: 26485516 DOI: 10.1039/c5mt00215j] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The kidney has recently emerged as an organ with a significant role in systemic iron (Fe) homeostasis. Substantial amounts of Fe are filtered by the kidney, which have to be reabsorbed to prevent Fe deficiency. Accordingly Fe transporters and receptors for protein-bound Fe are expressed in the nephron that may also function as entry pathways for toxic metals, such as cadmium (Cd), by way of "ionic and molecular mimicry". Similarities, but also differences in handling of Cd by these transport routes offer rationales for the propensity of the kidney to develop Cd toxicity. This critical review provides a comprehensive update on Fe transport by the kidney and its relevance for physiology and Cd nephrotoxicity. Based on quantitative considerations, we have also estimated the in vivo relevance of the described transport pathways for physiology and toxicology. Under physiological conditions all segments of the kidney tubules are likely to utilize Fe for cellular Fe requiring processes for metabolic purposes and also to contribute to reabsorption of free and bound forms of Fe into the circulation. But Cd entering tubule cells disrupts metabolic pathways and is unable to exit. Furthermore, our quantitative analyses contest established models linking chronic Cd nephrotoxicity to proximal tubular uptake of metallothionein-bound Cd. Hence, Fe transport by the kidney may be beneficial by preventing losses from the body. But increased uptake of Fe or Cd that cannot exit tubule cells may lead to kidney injury, and Fe deficiency may facilitate renal Cd uptake.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute of Physiology, Pathophysiology & Toxicology, Center for Biomedical Training and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 12, 58453 Witten, Germany.
| | - Natascha A Wolff
- Institute of Physiology, Pathophysiology & Toxicology, Center for Biomedical Training and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 12, 58453 Witten, Germany.
| |
Collapse
|
18
|
Sun MY, Woolley JC, Blohowiak SE, Smith ZR, Siddappa AM, Magness RR, Kling PJ. Dietary-induced gestational iron deficiency inhibits postnatal tissue iron delivery and postpones the cessation of active nephrogenesis in rats. Reprod Fertil Dev 2016; 29:RD15358. [PMID: 26876724 PMCID: PMC5577434 DOI: 10.1071/rd15358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/14/2015] [Indexed: 02/01/2023] Open
Abstract
Gestational iron deficiency (ID) can alter developmental programming through impaired nephron endowment, leading to adult hypertension, but nephrogenesis is unstudied. Iron status and renal development during dietary-induced gestational ID (<6 mg Fe kg-1 diet from Gestational Day 2 to Postnatal Day (PND) 7) were compared with control rats (198 mg Fe kg-1 diet). On PND2-PND10, PND15, PND30 and PND45, blood and tissue iron status were assessed. Nephrogenic zone maturation (PND2-PND10), radial glomerular counts (RGCs), glomerular size density and total planar surface area (PND15 and PND30) were also assessed. Blood pressure (BP) was measured in offspring. ID rats were smaller, exhibiting lower erythrocyte and tissue iron than control rats (PND2-PND10), but these parameters returned to control values by PND30-PND45. Relative kidney iron (µg g-1 wet weight) at PND2-PND10 was directly related to transport iron measures. In ID rats, the maturation of the active nephrogenic zone was later than control. RGCs, glomerular size, glomerular density, and glomerular planar surface area were lower than control at PND15, but returned to control by PND30. After weaning, the kidney weight/rat weight ratio (mg g-1) was heavier in ID than control rats. BP readings at PND45 were lower in ID than control rats. Altered kidney maturation and renal adaptations may contribute to glomerular size, early hyperfiltration and long-term renal function.
Collapse
Affiliation(s)
- Mary Y. Sun
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
- Obstetrics and Gynecology Perinatal Research Laboratories, University of Wisconsin, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Joseph C. Woolley
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Sharon E. Blohowiak
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Zachary R. Smith
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Ashajyothi M. Siddappa
- Department of Pediatrics, Division of Neonatology, Hennepin County Medical Center and University of Minnesota, Minneapolis, MN, USA
- Center for Neurobehavioral Development, 516 Delaware St. SE, Minneapolis, MN 55455, USA
| | - Ronald R. Magness
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
- Obstetrics and Gynecology Perinatal Research Laboratories, University of Wisconsin, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Pamela J. Kling
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| |
Collapse
|
19
|
Menon AV, Chang J, Kim J. Mechanisms of divalent metal toxicity in affective disorders. Toxicology 2015; 339:58-72. [PMID: 26551072 DOI: 10.1016/j.tox.2015.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/19/2015] [Accepted: 11/03/2015] [Indexed: 01/01/2023]
Abstract
Metals are required for proper brain development and play an important role in a number of neurobiological functions. The divalent metal transporter 1 (DMT1) is a major metal transporter involved in the absorption and metabolism of several essential metals like iron and manganese. However, non-essential divalent metals are also transported through this transporter. Therefore, altered expression of DMT1 can modify the absorption of toxic metals and metal-induced toxicity. An accumulating body of evidence has suggested that increased metal stores in the brain are associated with elevated oxidative stress promoted by the ability of metals to catalyze redox reactions, resulting in abnormal neurobehavioral function and the progression of neurodegenerative diseases. Metal overload has also been implicated in impaired emotional behavior, although the underlying mechanisms are not well understood with limited information. The current review focuses on psychiatric dysfunction associated with imbalanced metabolism of metals that are transported by DMT1. The investigations with respect to the toxic effects of metal overload on behavior and their underlying mechanisms of toxicity could provide several new therapeutic targets to treat metal-associated affective disorders.
Collapse
Affiliation(s)
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Mukaide T, Hattori Y, Misawa N, Funahashi S, Jiang L, Hirayama T, Nagasawa H, Toyokuni S. Histological detection of catalytic ferrous iron with the selective turn-on fluorescent probe RhoNox-1 in a Fenton reaction-based rat renal carcinogenesis model. Free Radic Res 2014; 48:990-5. [DOI: 10.3109/10715762.2014.898844] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
21
|
Veuthey T, Wessling-Resnick M. Pathophysiology of the Belgrade rat. Front Pharmacol 2014; 5:82. [PMID: 24795636 PMCID: PMC4000996 DOI: 10.3389/fphar.2014.00082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/02/2014] [Indexed: 01/01/2023] Open
Abstract
The Belgrade rat is an animal model of divalent metal transporter 1 (DMT1) deficiency. This strain originates from an X-irradiation experiment first reported in 1966. Since then, the Belgrade rat’s pathophysiology has helped to reveal the importance of iron balance and the role of DMT1. This review discusses our current understanding of iron transport homeostasis and summarizes molecular details of DMT1 function. We describe how studies of the Belgrade rat have revealed key roles for DMT1 in iron distribution to red blood cells as well as duodenal iron absorption. The Belgrade rat’s pathology has extended our knowledge of hepatic iron handling, pulmonary and olfactory iron transport as well as brain iron uptake and renal iron handling. For example, relationships between iron and manganese metabolism have been discerned since both are essential metals transported by DMT1. Pathophysiologic features of the Belgrade rat provide us with a unique and interesting animal model to understand iron homeostasis.
Collapse
Affiliation(s)
- Tania Veuthey
- Department of Genetics and Complex Diseases, Harvard School of Public Health Boston, MA, USA
| | | |
Collapse
|