1
|
Rajkowska-Myśliwiec M, Szczuko M, Witczak A, Kaczkan M, Małgorzewicz S. Assessment of essential and toxic trace element levels in erythrocytes of hemodialysis patients with end-stage renal disease. J Trace Elem Med Biol 2024; 85:127491. [PMID: 38943837 DOI: 10.1016/j.jtemb.2024.127491] [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: 05/02/2024] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a global public health problem, resulting in end-stage kidney disease, cardiovascular disease, and premature death. AIM The aim of the study was to determine the profile of essential and toxic trace elements in erythrocytes of patients with end-stage renal disease (ESRD) and their relationship with selected anthropometric and biochemical parameters. METHODS The present study compared the profiles of trace elements, including toxic sub-stances, in the erythrocytes of 80 hemodialysis patients with CKD with 40 healthy subjects. All patients had stage 5 CKD. The levels of Cd and Pb were determined by graphite furnace atomic absorption spectrometry and levels of Fe, Mn, Zn, Cu Cr, Ni, and Li by inductively coupled plasma atomic emission spectrometry. RESULTS The ESRD patients demonstrated significantly lower Fe and Zn concentrations and significantly higher Mn and Li and toxic Pb and Cd concentrations in erythrocytes compared to those of the healthy controls. Negative correlations were observed, among others, between the concentrations of Cu, Li, and creatinine; Cu and phosphates; Mn, Pb, and transferrin saturation while positive correlations were noted between Cu, Cr, and transferrin and Pb, Cr, and the normalized protein catabolism rate. CONCLUSIONS The higher concentrations of toxic elements present in the erythrocytes of CKD patients might have resulted from the reduced ability of the kidneys to excrete them. Moreover, differences in the concentrations of essential elements (Fe, Mn, Zn) between the two groups indicated that their resorption in the kidneys of CKD patients was impaired. Patients with CKD might benefit from interventions intended to reduce high, toxic concentrations of Pb and Cd and Li and Mn as an alternative supportive treatment. Iron and zinc supplementation should be a component for the treatment of anemia in CKD patients.
Collapse
Affiliation(s)
- Monika Rajkowska-Myśliwiec
- Department of Toxicology, Dairy Technology and Food Storage, Faculty of Food Science and Fisheries, West Pomeranian University of Technology in Szczecin, Papieza Pawla VI 3, Szczecin 71-459, Poland.
| | - Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Władysława Broniewskiego 24, Szczecin 71-460, Poland.
| | - Agata Witczak
- Department of Toxicology, Dairy Technology and Food Storage, Faculty of Food Science and Fisheries, West Pomeranian University of Technology in Szczecin, Papieza Pawla VI 3, Szczecin 71-459, Poland
| | - Małgorzata Kaczkan
- Department of Clinical Nutrition, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, Gdańsk 80-211, Poland
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, Gdańsk 80-211, Poland
| |
Collapse
|
2
|
Ma X, Lv J, Zhang S, Zhang X, Lin X, Li S, Yang L, Xue F, Yi F, Zhang T. Habitual Iron Supplementation Associated with Elevated Risk of Chronic Kidney Disease in Individuals with Antihypertensive Medication. Nutrients 2024; 16:2355. [PMID: 39064798 PMCID: PMC11279651 DOI: 10.3390/nu16142355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The aim of this study was to examine the effects of habitual iron supplementation on the risk of CKD in individuals with different hypertensive statuses and antihypertension treatment statuses. We included a total of 427,939 participants in the UK Biobank study, who were free of CKD and with complete data on blood pressure at baseline. Cox proportional hazards regression models were used to examine the adjusted hazard ratios of habitual iron supplementation for CKD risk. After multivariable adjustment, habitual iron supplementation was found to be associated with a significantly higher risk of incident CKD in hypertensive participants (HR 1.12, 95% CI 1.02 to 1.22), particularly in those using antihypertensive medication (HR 1.21, 95% CI 1.08 to 1.35). In contrast, there was no significant association either in normotensive participants (HR 1.06, 95% CI 0.94 to 1.20) or in hypertensive participants without antihypertensive medication (HR 1.02, 95% CI 0.90 to 1.17). Consistently, significant multiplicative and additive interactions were observed between habitual iron supplementation and antihypertensive medication on the risk of incident CKD (p all interaction < 0.05). In conclusion, habitual iron supplementation was related to a higher risk of incident CKD among hypertensive patients, the association might be driven by the use of antihypertensive medication.
Collapse
Affiliation(s)
- Xiaoyan Ma
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jiali Lv
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shuai Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaofeng Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xia Lin
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shengxu Li
- Children’s Minnesota Research Institute, Children’s Minnesota, Minneapolis, MN 55404, USA;
| | - Lin Yang
- Department of Cancer Epidemiology and Prevention Research, Cancer Research & Analytics, Cancer Care Alberta, Alberta Health Services, Calgary, AB T2V 0N5, Canada;
- Departments of Oncology and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2V 0N5, Canada
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (X.M.); (J.L.); (S.Z.); (X.Z.); (X.L.); (F.X.)
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
3
|
Ali Fadlalmola H, Al-Sayaghi KM, Al-Hebshi AA, Alhujaily M, Alyamani AO, Alem AA, Syrafi MH, Alem S, Farhat AH, Mohamed FA, Abdalrahman HH, Abdelmalik MA, Abdalrhman NM, Eltayeb AM. Efficacy of daprodustat for patients on dialysis with anemia: systematic review and network meta-analysis. Pan Afr Med J 2024; 47:114. [PMID: 38828426 PMCID: PMC11143073 DOI: 10.11604/pamj.2024.47.114.37278] [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: 09/09/2022] [Accepted: 03/05/2024] [Indexed: 06/05/2024] Open
Abstract
Chronic kidney disease (CKD) is commonly complicated by anemia. Treating dialysis-dependent patients with anemia, including daprodustat and other inhibitors of prolyl hydroxylase of hypoxia-inducible factor, recombinant human erythropoietin (rhEPO), and iron supplements. We conducted this study to test our postulation; daprodustat is superior to rhEPO and other conventional treatments respecting efficacy and safety parameters. We made systematic search through PubMed, Web of Science, Scopus, and Cochrane. Seven unique trials were eventually included for systematic review; six of them with a sample size of 759 patients entered our network meta-analysis (NMA). Daprodustat 25-30 mg was associated with the greatest change in serum hemoglobin (MD=1.86, 95%CI= [1.20; 2.52]), ferritin (MD= -180.84, 95%CI= [-264.47; -97.20]), and total iron binding capacity (TIBC) (MD=11.03, 95%CI= [3.15; 18.92]) from baseline values. Dialysis-dependent patients with anemia had a significant increment in serum Hemoglobin and TIBC and a reduction in serum ferritin, in a dose-dependent manner, when administered daprodustat.
Collapse
Affiliation(s)
- Hammad Ali Fadlalmola
- Department of Community Health Nursing, College of Nursing, Taibah University, Al-Madinah Al-Munawarah, Kingdom of Saudi Arabia
| | - Khaled Mohammed Al-Sayaghi
- Department of Medical Surgical Nursing, College of Nursing, Taibah University, Al-Madinah Al-Munawarah, Kingdom of Saudi Arabia
- Nursing Division, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Yemen
| | - Abdulqader Abdlah Al-Hebshi
- Pediatric Hematology Oncology Service, Department of Pediatrics, Prince Mohammed Bin AbdulAziz Hospital, Ministry of National Guard Health Affairs, Medina, Saudi Arabia
| | - Muhanad Alhujaily
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Arwa Omar Alyamani
- Service of Pediatric Hematology Oncology, Department of Pediatrics, King Saud Bin Abdul Azizi Collage for Health Science, Riyadh, Saudi Arabia
- Princess Noorah Oncology Center, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Alaa Abdulrhman Alem
- Nephrology Service, Department of Medicine, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Mona Hamza Syrafi
- Service of Pediatric Nephrology, Department of Pediatrics, King Salman Bin Abdulaziz Medical City, Medina, Saudi Arabia
| | - Sarah Alem
- Nahdi Medical Company, Medina, Saudi Arabia
| | - Afrah Hassan Farhat
- Department of Pediatrics, King Khalid University Hospital, Riyadh, Saudi Arabia
| | - Fathi Abdelrazig Mohamed
- Department of Pediatrics, Prince Mohammed Bin AbdulAziz Hospital, Ministry of National Guard Health Affairs, Medina, Saudi Arabia
| | | | - Mohammed Abdelkrim Abdelmalik
- Department of Nursing, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
- Faculty of Medicine and Health Sciences, Nursing, University of El Imam El Mahdi Kosti, White Nile, Sudan
| | | | | |
Collapse
|
4
|
Punchai S, Chaiyagot N, Artkaew N, Jusakul A, Cha’on U, Thanan R, Vaeteewoottacharn K, Lert-Itthiporn W. Iron-induced kidney cell damage: insights into molecular mechanisms and potential diagnostic significance of urinary FTL. Front Mol Biosci 2024; 11:1352032. [PMID: 38449697 PMCID: PMC10916690 DOI: 10.3389/fmolb.2024.1352032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
Background: Iron overload can lead to organ and cell injuries. Although the mechanisms of iron-induced cell damage have been extensively studied using various cells, little is known about these processes in kidney cells. Methods: In this study, we first examined the correlation between serum iron levels and kidney function. Subsequently, we investigated the molecular impact of excess iron on kidney cell lines, HEK293T and HK-2. The presence of the upregulated protein was further validated in urine. Results: The results revealed that excess iron caused significant cell death accompanied by morphological changes. Transcriptomic analysis revealed an up-regulation of the ferroptosis pathway during iron treatment. This was confirmed by up-regulation of ferroptosis markers, ferritin light chain (FTL), and prostaglandin-endoperoxide synthase 2 (PTGS2), and down-regulation of acyl-CoA synthetase long-chain family member 4 (ACSL4) and glutathione peroxidase 4 (GPX4) using real-time PCR and Western blotting. In addition, excess iron treatment enhanced protein and lipid oxidation. Supportively, an inverse correlation between urinary FTL protein level and kidney function was observed. Conclusion: These findings suggest that excess iron disrupts cellular homeostasis and affects key proteins involved in kidney cell death. Our study demonstrated that high iron levels caused kidney cell damage. Additionally, urinary FTL might be a useful biomarker to detect kidney damage caused by iron toxicity. Our study also provided insights into the molecular mechanisms of iron-induced kidney injury, discussing several potential targets for future interventions.
Collapse
Affiliation(s)
- Soraya Punchai
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nachayada Chaiyagot
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nadthanicha Artkaew
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Chronic Kidney Disease Prevention in Northeastern Thailand, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Chronic Kidney Disease Prevention in Northeastern Thailand, Khon Kaen University, Khon Kaen, Thailand
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Ubon Cha’on
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Chronic Kidney Disease Prevention in Northeastern Thailand, Khon Kaen University, Khon Kaen, Thailand
| | - Raynoo Thanan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Chronic Kidney Disease Prevention in Northeastern Thailand, Khon Kaen University, Khon Kaen, Thailand
| | - Kulthida Vaeteewoottacharn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Worachart Lert-Itthiporn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Chronic Kidney Disease Prevention in Northeastern Thailand, Khon Kaen University, Khon Kaen, Thailand
- Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
5
|
Shrestha P, Paul S, Sumida K, Thomas F, Surbhi S, Naser AM, Streja E, Rhee CM, Kalantar-Zadeh K, Kovesdy CP. Association of iron therapy with incidence of chronic kidney disease. Eur J Haematol 2023; 111:872-880. [PMID: 37668586 DOI: 10.1111/ejh.14091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
OBJECTIVE We investigated the association of oral iron replacement with the incidence of chronic kidney disease (CKD) in a population with normal kidney function to study the effects of iron replacement on the development of new onset CKD. METHODS In a national cohort of US Veterans with no pre-existing CKD, we identified 33 894 incident new users of oral iron replacement and a comparable group of 112 780 patients who did not receive any iron replacement during 2004-2018. We examined the association of oral iron replacement versus no iron replacement with the incidence of eGFR <60 mL/min/1.73 m2 and the incidence of urine albumin creatinine ratio (UACR) ≥30 mg/g in competing risk regressions and in Cox models. We used propensity score weighing to account for differences in key baseline characteristics associated with the use of oral iron replacement. RESULTS In the cohort of 146 674 patients, a total of 18 547 (13%) patients experienced incident eGFR <60 mL/min/1.73 m2 , and 16 117 patients (11%) experienced new onset UACR ≥30 mg/g. Oral iron replacement was associated with significantly higher risk of incident eGFR <60 mL/min/1.73 m2 (subhazard ratio, 95% confidence interval [CI]: 1.3 [1.22-1.38], p < .001) and incident albuminuria (subhazard ratio, 95% CI: 1.14 [1.07-1.22], p < .001). CONCLUSION Oral iron replacement is associated with higher risk of new onset CKD. The long-term kidney safety of oral iron replacement should be tested in clinical trials.
Collapse
Affiliation(s)
- Prabin Shrestha
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Shejuti Paul
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Keiichi Sumida
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Fridtjof Thomas
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Satya Surbhi
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Abu Mohd Naser
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, Tennessee, USA
| | - Elani Streja
- Division of Nephrology and Hypertension, University of California-Irvine, Orange, California, USA
| | - Connie M Rhee
- Division of Nephrology and Hypertension, University of California-Irvine, Orange, California, USA
- Long Beach VA Medical Center, Long Beach, California, USA
| | - Kamyar Kalantar-Zadeh
- Long Beach VA Medical Center, Long Beach, California, USA
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension, Habor-UCLA Medical Center and the Lundquist Institute, Torrance, California, USA
| | - Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Nephrology Section, Memphis VA Medical Center, Memphis, Tennessee, USA
| |
Collapse
|
6
|
Anne Marie U, Murererehe J, Rehman M, Chittilla M, Uwambaye P, Razzaque MS. Oral manifestations of iron imbalance. Front Nutr 2023; 10:1272902. [PMID: 37899821 PMCID: PMC10611504 DOI: 10.3389/fnut.2023.1272902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/11/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Uwitonze Anne Marie
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
| | - Julienne Murererehe
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
| | - Mahum Rehman
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Mythri Chittilla
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Peace Uwambaye
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
| | - Mohammed S. Razzaque
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| |
Collapse
|
7
|
Shaddinger B, Mahar KM, Sprys M, Andrews SM, Chattoraj S, Israni R, Cobitz A. Comparison of Two Manufacturing Processes of Daprodustat for Bioequivalence and Dissolution in Healthy Volunteers: A Randomized Crossover Study. Clin Pharmacol Drug Dev 2023; 12:739-748. [PMID: 37125459 DOI: 10.1002/cpdd.1257] [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: 11/15/2022] [Accepted: 03/13/2023] [Indexed: 05/02/2023]
Abstract
Daprodustat, an orally bioavailable hypoxia-inducible factor-prolyl hydroxylase enzyme inhibitor, has recently completed phase 3 clinical development for treating anemia of chronic kidney disease. Part A of this 2-part, randomized, double-blind, single-dose, cross-over study (NCT04640311) compared pharmacokinetic properties of a single oral dose of daprodustat 4 mg tablets manufactured via twin-screw wet granulation (process 1) to 2 sets of 4 mg tablets manufactured via high-shear wet granulation (process 2), to assess the impact of different dissolution profiles on pharmacokinetics. Part B assessed the bioequivalence of daprodustat tablets manufactured via process 1 with tablets manufactured via process 2 at 5 different dose strengths (1, 2, 4, 6, and 8 mg). In part A, mean plasma concentrations of daprodustat were comparable over a 24-hour period despite differences in manufacturing processes and dissolution profiles. In part B, the 90% confidence intervals of the ratios of the least squared means for area under the concentration-time curve and maximum observed plasma concentration fell within the 0.8-1.25 bioequivalence range for all doses, except for maximum observed plasma concentration at 8 mg. A prespecified sensitivity analysis jointly assessing all doses showed bioequivalence for all doses tested. No new safety concerns for daprodustat were identified.
Collapse
Affiliation(s)
| | | | | | - Susan M Andrews
- Global Clinical Operations Development R&D, GSK, Research Triangle Park, Durham, North Carolina, USA
| | | | | | | |
Collapse
|
8
|
Harlow CE, Patel VV, Waterworth DM, Wood AR, Beaumont RN, Ruth KS, Tyrrell J, Oguro-Ando A, Chu AY, Frayling TM. Genetically proxied therapeutic prolyl-hydroxylase inhibition and cardiovascular risk. Hum Mol Genet 2023; 32:496-505. [PMID: 36048866 PMCID: PMC9851745 DOI: 10.1093/hmg/ddac215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/05/2022] [Accepted: 08/22/2022] [Indexed: 01/24/2023] Open
Abstract
Prolyl hydroxylase (PHD) inhibitors are in clinical development for anaemia in chronic kidney disease. Epidemiological studies have reported conflicting results regarding safety of long-term therapeutic haemoglobin (Hgb) rises through PHD inhibition on risk of cardiovascular disease. Genetic variation in genes encoding PHDs can be used as partial proxies to investigate the potential effects of long-term Hgb rises. We used Mendelian randomization to investigate the effect of long-term Hgb level rises through genetically proxied PHD inhibition on coronary artery disease (CAD: 60 801 cases; 123 504 controls), myocardial infarction (MI: 42 561 cases; 123 504 controls) or stroke (40 585 cases; 406 111 controls). To further characterize long-term effects of Hgb level rises, we performed a phenome-wide association study (PheWAS) in up to 451 099 UK Biobank individuals. Genetically proxied therapeutic PHD inhibition, equivalent to a 1.00 g/dl increase in Hgb levels, was not associated (at P < 0.05) with increased odds of CAD; odd ratio (OR) [95% confidence intervals (CI)] = 1.06 (0.84, 1.35), MI [OR (95% CI) = 1.02 (0.79, 1.33)] or stroke [OR (95% CI) = 0.91 (0.66, 1.24)]. PheWAS revealed associations with blood related phenotypes consistent with EGLN's role, relevant kidney- and liver-related biomarkers like estimated glomerular filtration rate and microalbuminuria, and non-alcoholic fatty liver disease (Bonferroni-adjusted P < 5.42E-05) but these were not clinically meaningful. These findings suggest that long-term alterations in Hgb through PHD inhibition are unlikely to substantially increase cardiovascular disease risk; using large disease genome-wide association study data, we could exclude ORs of 1.35 for cardiovascular risk with a 1.00 g/dl increase in Hgb.
Collapse
Affiliation(s)
- Charli E Harlow
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| | - Vickas V Patel
- GlaxoSmithKline, Collegeville, PA 19426, USA
- Spark Therapeutics, Inc., Philadelphia, PA 19104, USA
| | - Dawn M Waterworth
- GlaxoSmithKline, Collegeville, PA 19426, USA
- Immunology Translational Sciences, Janssen, Spring House, PA 19044, USA
| | - Andrew R Wood
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| | - Robin N Beaumont
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| | - Katherine S Ruth
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| | - Jessica Tyrrell
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| | - Asami Oguro-Ando
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| | | | - Timothy M Frayling
- College of Medicine and Health, University of Exeter, Exeter, Devon EX2 5DW, UK
| |
Collapse
|
9
|
Babker A, Talawy T, Bylappa S, Ismail M, Kandakurti P, Gopakumar A. Correlation of Erythroferrone and Hepcidin Hormones with Iron Status Levels in Patients with Iron Deficiency. JOURNAL OF APPLIED HEMATOLOGY 2023. [DOI: 10.4103/joah.joah_63_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
|
10
|
Xie Y, Liu F, Zhang X, Jin Y, Li Q, Shen H, Fu H, Mao J. Benefits and risks of essential trace elements in chronic kidney disease: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1400. [PMID: 36660676 PMCID: PMC9843383 DOI: 10.21037/atm-22-5969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023]
Abstract
Background and Objective Chronic kidney disease (CKD) is an important public health concern. With the decline of renal function, CKD patients gradually progress to end-stage kidney disease and need to undergo dialysis or kidney transplantation to maintain life, bringing a heavy economic burden to the family and society. Therefore, it is necessary to effectively prevent and delay the progression of CKD. Essential trace elements play an indispensable role in CKD, and the objective of this study is to systematically review their benefits in the disease and summarize the risks of their excess. Methods The keywords "trace elements", "chronic kidney disease", "dialysis", "inflammation", and "fibrosis" and their combinations were used to search for relevant literature published in the PubMed database and Web of Science. We then summarized the role of trace element abnormalities in CKD patients in anemia, oxidative stress, inflammation, and chronic fibrosis, and the risk of their excess. Key Content and Findings Imbalance of essential trace elements is a common complication of CKD and a risk factor for CKD progression, cardiovascular events, and death. This article reviews the effects of essential trace elements (iron, zinc, selenium, copper, iodine, and manganese) on CKD. We analyze literature and discuss the advantages and disadvantages of various essential trace elements. Conclusions Research shows CKD patients have an imbalance of essential trace elements, and treatment based on these is an important direction for future exploration. A knowledge of the homeostasis of trace elements is important to improving the prognosis of CKD patients and delaying the progression of the disease.
Collapse
Affiliation(s)
- Yi Xie
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Fei Liu
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaojing Zhang
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yanyan Jin
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiuyu Li
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Huijun Shen
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Haidong Fu
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
11
|
Perrone V, Veronesi C, Dovizio M, Ancona DD, Bartolini F, Ferrante F, Lupi A, Palcic S, Re D, Terlizzi AP, Ramirez de Arellano Serna A, Cogliati P, Degli Esposti L. The Influence of Iron-Deficiency Anaemia (IDA) Therapy on Clinical Outcomes and Healthcare Resource Consumptions in Chronic Kidney Disease Patients Affected by IDA: A Real-Word Evidence Study among the Italian Population. J Clin Med 2022; 11:jcm11195820. [PMID: 36233688 PMCID: PMC9573467 DOI: 10.3390/jcm11195820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Anaemia is a uraemia-related complication frequently found in non-dialysis-dependent chronic kidney disease (ND-CKD) patients, with iron-deficiency anaemia (IDA) as the main underlying mechanism. Given the suboptimal anaemia management in ND-CKD patients with a co-diagnosis of IDA, this study evaluated the role of IDA therapy on clinical outcomes and healthcare resource consumptions in an Italian clinical setting. A retrospective observational real-world analysis was performed on administrative databases of healthcare entities, covering around 6.9 million health-assisted individuals. From January 2010 to March 2019, ND-CKD patients were included and diagnosed with IDA in the presence of two low-haemoglobin (Hb) measurements. Patients were divided into IDA-treated and untreated, based on the prescription of iron [Anatomical-Therapeutic Chemical (ATC) code B03A] or anti-anaemia preparations (ATC code B03X), and evaluated during a 6-month follow-up from the index date [first low haemoglobin (Hb) detection]. IDA treatment resulted in significantly decreased incidence of all cause-related, cardiovascular-related, and IDA-related hospitalizations (treated vs. untreated: 44.5% vs. 81.8%, 12.3% vs. 25.3%, and 16.2% vs. 26.2%, respectively, p < 0.001). A healthcare direct cost estimation showed that overall mean expenditure per patient reduced by 47% with IDA treatment (5245€ vs. 9918€, p < 0.001), mainly attributable to hospitalizations (3767€ vs. 8486€, p < 0.001). This real-life analysis on Italian ND-CKD-IDA patients indicates that IDA therapy administration provides significant benefits in terms of patients’ clinical outcomes and healthcare cost savings.
Collapse
Affiliation(s)
- Valentina Perrone
- CliCon Società Benefit S.r.l. Health, Economics & Outcomes Research, 40137 Bologna, Italy
| | - Chiara Veronesi
- CliCon Società Benefit S.r.l. Health, Economics & Outcomes Research, 40137 Bologna, Italy
| | - Melania Dovizio
- CliCon Società Benefit S.r.l. Health, Economics & Outcomes Research, 40137 Bologna, Italy
| | - Domenica Daniela Ancona
- Dipartimento Farmaceutico, Azienda Sanitaria Locale delle province di Barletta- Andria- Trani (BAT), 76125 Andria, Italy
| | - Fausto Bartolini
- Dipartimento Farmaceutico, Unità Sanitaria Locale Umbria 2, 05100 Terni, Italy
| | - Fulvio Ferrante
- Dipartimento della Diagnostica ed Assistenza Farmaceutica, ASL Frosinone, 03100 Frosinone, Italy
| | - Alessandro Lupi
- Azienda Sanitaria Locale del Verbano Cusio Ossola (VCO), 28887 Omegna, Italy
| | - Stefano Palcic
- Azienda Sanitaria Universitaria Integrata Giuliano-Isontina (ASUGI), 34148 Trieste, Italy
| | - Davide Re
- Dipartimento Assistenza Territoriale, Azienda Sanitaria Locale di Teramo, 64100 Teramo, Italy
| | - Annamaria Pia Terlizzi
- Dipartimento Farmaceutico, Azienda Sanitaria Locale delle province di Barletta- Andria- Trani (BAT), 76125 Andria, Italy
| | | | | | - Luca Degli Esposti
- CliCon Società Benefit S.r.l. Health, Economics & Outcomes Research, 40137 Bologna, Italy
- Correspondence:
| |
Collapse
|
12
|
Fadlalmola H, Al-Sayaghi K, Al-Hebshi A, Aljohani M, Albalawi M, Kashari O, Alem A, Alrasheedy M, Balelah S, Almuteri F, Alyamani A, Alwasaidi T. Efficacy of Different Doses of Daprodustat for Anemic Non-dialysis Patients with Chronic Kidney Disease: A Systematic Review and Network Meta-Analysis. J Clin Med 2022; 11:2722. [PMID: 35628849 PMCID: PMC9145143 DOI: 10.3390/jcm11102722] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/28/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
(1) Background: Anemia affects about 40% of patients with chronic kidney disease (CKD). Daprodustat improves serum hemoglobin in anemic patients by inhibiting prolyl hydroxylase of hypoxia-inducible factor. We conducted a network meta-analysis to investigate the direct and indirect effects of different doses of daprodustat compared to each other and erythropoietin and placebo. (2) Methods: We searched PubMed, Cochrane Library, Web of Science, and Scopus, for randomized clinical trials (RCTs) reporting data about different doses of daprodustat for anemia in nondialysis of CKDs. (3) Results: We eventually included five RCTs with a total sample size of 4566 patients. We found that the higher the dose of daprodustat, the greater the change in serum total iron binding capacity (TIBC), hemoglobin, and ferritin from baseline. Compared to placebo, daprodustat 25-30 mg was associated with the highest significant increase in serum hemoglobin (MD = 3.27, 95% CI = [1.89; 4.65]), a decrease in serum ferritin (MD = -241.77, 95% CI = [-365.45; -118.09]) and increase in serum TIBC (MD = 18.52, 95% CI = [12.17; 24.87]). (4) Conclusion: Higher daprodustat doses were associated with a higher impact on efficacy outcomes as serum total iron-binding capacity (TIBC), hemoglobin, and ferritin. However, data about the safety profile of different doses of daprodustat is still missing.
Collapse
Affiliation(s)
- Hammad Fadlalmola
- Department of Community Health Nursing, College of Nursing, Taibah University, Al-Madinah Al-Munawarah and KSA 1, Medina 42353, Saudi Arabia
| | - Khaled Al-Sayaghi
- Department of Medical Surgical Nursing, College of Nursing, Taibah University, Al-Madinah Al-Munawarah, KSA 1, Medina 42353, Saudi Arabia;
- Nursing Division, Faculty of Medicine and Health Sciences, Sana’a University, Sana’a P.O. Box 1247, Yemen
| | - Abdulqader Al-Hebshi
- Department of Pediatrics, Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, Medina 42353, Saudi Arabia;
| | - Maher Aljohani
- Department of Pathology, College of Medicine, Taibah University, Medina 42353, Saudi Arabia;
- Department of Pathology and Laboratory Medicine, Prince Mohammed Bin Abdulaziz Hospital, Medina 42353, Saudi Arabia
| | - Mohammed Albalawi
- Department of Medicine, College of Medicine, Taibah University, Medina 42353, Saudi Arabia; (M.A.); (A.A.); (T.A.)
| | - Ohoud Kashari
- Al Aziziyah Children Hospital, Jeddah 22253, Saudi Arabia;
| | - Alaa Alem
- Department of Medicine, College of Medicine, Taibah University, Medina 42353, Saudi Arabia; (M.A.); (A.A.); (T.A.)
| | - Mariam Alrasheedy
- Department of Pediatrics, East Jeddah Hospital, Jeddah 22253, Saudi Arabia;
| | - Saud Balelah
- Department of Medicine, King Fahad Hospital, Medina 42353, Saudi Arabia;
| | - Faten Almuteri
- Department of Pediatrics, King Salman Bin Abdulaziz Medical City, Madinah 42353, Saudi Arabia;
| | - Arwa Alyamani
- Department of Oncology, King Saud Bin Abdulaziz Collage for Health Science, Jeddah 22253, Saudi Arabia; or
- Princess Noorah Oncology Center, Ministry of National Guard Health Affairs, Jeddah 22253, Saudi Arabia
| | - Turki Alwasaidi
- Department of Medicine, College of Medicine, Taibah University, Medina 42353, Saudi Arabia; (M.A.); (A.A.); (T.A.)
- Prince Mohammed Bin Abdulaziz Hospital, Ministry of National Guard Health Affairs, Medina 42353, Saudi Arabia
| |
Collapse
|
13
|
Evaluation of Predicting the Value of the Reticulocyte Hemoglobin Equivalent for Iron Deficiency in Chronic Kidney Disease Patients. Nephrourol Mon 2022. [DOI: 10.5812/numonthly-121289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background: Iron management is essential for anemia treatment in chronic kidney disease. The reticulocyte hemoglobin equivalent (RET-He) is a reticulocyte parameter that reflects hemoglobin synthesis of newly formed erythrocytes in the bone marrow in real-time. Objectives: This study aims to evaluate the role of reticulocyte hemoglobin equivalent (RET-He) in predicting iron deficiency in chronic kidney disease (CKD) patients. Methods: Following a descriptive cross-sectional observational design, this study was conducted on 131 adult patients with CKD stages 3 - 5. Laboratory indices, including complete blood count, some biochemical indices, iron status, and reticulocyte indices (including RET-He), were measured. Iron deficiency (ID) was defined as TSAT < 20%, where serum ferritin level > 100 ng/mL was defined as functional ID, while serum ferritin level
Collapse
|
14
|
Mahar KM, Shaddinger BC, Ramanjineyulu B, Andrews S, Caltabiano S, Lindsay AC, Cobitz AR. Pharmacokinetics of Daprodustat and Metabolites in Individuals with Normal and Impaired Hepatic Function. Clin Pharmacol Drug Dev 2022; 11:562-575. [PMID: 35355447 PMCID: PMC9310628 DOI: 10.1002/cpdd.1090] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/17/2022] [Indexed: 11/23/2022]
Abstract
Daprodustat is a hypoxia‐inducible factor‐prolyl hydroxylase inhibitor in development for treatment of anemia of chronic kidney disease. We evaluated the role of hepatic impairment on daprodustat pharmacokinetics, pharmacodynamics, and tolerability. Participants with mild (Child‐Pugh Class A, score 5‒6) and moderate (Child‐Pugh Class B, score 7‒9) hepatic impairment and matched healthy controls were administered single 6‐mg doses of daprodustat. Exposure parameters were determined for daprodustat and its six metabolites. Comparisons resulted in 1.5‐ and 2.0‐fold higher daprodustat Cmax and area under the curve (AUC) exposures in participants with mild and moderate hepatic impairment, respectively, versus controls; Cmax in mild hepatic impairment was comparable to controls. Similarly, aligned with parent drug, unbound daprodustat Cmax and AUC exposures increased 1.6‐ to 2.3‐fold in hepatic‐impaired participants versus controls, and metabolite exposures were 1.2‐ to 2.0‐fold higher in participants with hepatic impairment. Erythropoeitin (EPO) baseline‐corrected AUC exposures were between 0.3‐fold lower and 2.2‐fold higher in matched controls versus hepatic‐impaired participants. No serious or study drug‐related adverse events were reported. Daprodustat exposure was increased in participants with moderate and mild hepatic impairment compared with matched controls; however, no meaningful differences in EPO were observed and no new safety concerns were identified (ClinicalTrials.gov: NCT03223337).
Collapse
Affiliation(s)
- Kelly M Mahar
- Clinical Pharmacology Modeling & Simulation, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | | | - Susan Andrews
- Clinical Science & Study Operations, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Stephen Caltabiano
- Medicine Delivery Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Alistair C Lindsay
- Medicine Delivery Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Alexander R Cobitz
- Medicine Delivery Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| |
Collapse
|
15
|
Garavaglia ML, Giustarini D, Colombo G, Reggiani F, Finazzi S, Calatroni M, Landoni L, Portinaro NM, Milzani A, Badalamenti S, Rossi R, Dalle-Donne I. Blood Thiol Redox State in Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23052853. [PMID: 35269995 PMCID: PMC8911004 DOI: 10.3390/ijms23052853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023] Open
Abstract
Thiols (sulfhydryl groups) are effective antioxidants that can preserve the correct structure of proteins, and can protect cells and tissues from damage induced by oxidative stress. Abnormal levels of thiols have been measured in the blood of patients with moderate-to-severe chronic kidney disease (CKD) compared to healthy subjects, as well as in end-stage renal disease (ESRD) patients on haemodialysis or peritoneal dialysis. The levels of protein thiols (a measure of the endogenous antioxidant capacity inversely related to protein oxidation) and S-thiolated proteins (mixed disulphides of protein thiols and low molecular mass thiols), and the protein thiolation index (the molar ratio of the S-thiolated proteins to free protein thiols in plasma) have been investigated in the plasma or red blood cells of CKD and ESRD patients as possible biomarkers of oxidative stress. This type of minimally invasive analysis provides valuable information on the redox status of the less-easily accessible tissues and organs, and of the whole organism. This review provides an overview of reversible modifications in protein thiols in the setting of CKD and renal replacement therapy. The evidence suggests that protein thiols, S-thiolated proteins, and the protein thiolation index are promising biomarkers of reversible oxidative stress that could be included in the routine monitoring of CKD and ESRD patients.
Collapse
Affiliation(s)
- Maria Lisa Garavaglia
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (M.L.G.); (G.C.); (L.L.); (A.M.)
| | - Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018–2022), University of Siena, Via A. Moro 2, 53100 Siena, Italy;
| | - Graziano Colombo
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (M.L.G.); (G.C.); (L.L.); (A.M.)
| | - Francesco Reggiani
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy or (F.R.); (S.F.); or (M.C.); (S.B.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Silvia Finazzi
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy or (F.R.); (S.F.); or (M.C.); (S.B.)
| | - Marta Calatroni
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy or (F.R.); (S.F.); or (M.C.); (S.B.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Lucia Landoni
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (M.L.G.); (G.C.); (L.L.); (A.M.)
| | - Nicola Marcello Portinaro
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Aldo Milzani
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (M.L.G.); (G.C.); (L.L.); (A.M.)
| | - Salvatore Badalamenti
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy or (F.R.); (S.F.); or (M.C.); (S.B.)
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018–2022), University of Siena, Via A. Moro 2, 53100 Siena, Italy;
- Correspondence: (R.R.); (I.D.-D.)
| | - Isabella Dalle-Donne
- Department of Biosciences (Department of Excellence 2018–2022), Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (M.L.G.); (G.C.); (L.L.); (A.M.)
- Correspondence: (R.R.); (I.D.-D.)
| |
Collapse
|
16
|
Zapora-Kurel A, Kuźma Ł, Zakrzewska M, Żórawski M, Dobrzycki S, Twardowska-Kawalec M, Małyszko J. Novel Iron Parameters in Patients with Type 2 Diabetes Mellitus in Relation to Kidney Function. J Clin Med 2021; 10:jcm10163732. [PMID: 34442028 PMCID: PMC8397038 DOI: 10.3390/jcm10163732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/AIMS Anemia of chronic disease is a common feature in diabetes and chronic kidney disease. Hepcidin is the key element involved in iron metabolism; however, studies on new indices of iron status are still ongoing. The aim of the study was to assess novel iron parameters in patients with type 2 diabetes mellitus in relation to kidney function. METHODS The study included 80 type 2 diabetic patients and 23 healthy volunteers. Standard laboratory measurements were used to measure the iron status, complete blood count, creatinine, the estimated glomerular filtration rate (eGFR), serum lipids, and brain natriuretic peptides (BNPs). Commercially available kits were used to measure hepcidin-25, the soluble transferrin receptor (sTfR), growth differentiation factor-15 (GDF-15), and hypoxia-inducible factor-1 alpha. RESULTS Anemia was present in 65% of the studied patients. The control group was found to have significantly higher hepcidin, sTfR, and GDF-15, and lower hemoglobin and iron. When compared with patients with eGFR values ≥60 mL/min/1.73 m2 and <60 mL/min/1.73 m2, we found that patients with higher eGFR had higher hemoglobin, ferritin, and HIF-1 alpha, lower BNP, and were younger. We found that levels of HIF-1 alpha are negligible in the studied population and were related to age only in patients with eGFR values ≥60 mL/min/1.73 m2. CONCLUSION A comprehensive assessment of iron status is rarely performed. Novel biomarkers of iron metabolism are not generally related to kidney function. Whether the assessment of HIF-1 alpha would be a marker of efficient anemia therapy with HIF-prolyl hydroxylase inhibitors is still a matter for further study.
Collapse
Affiliation(s)
- Agnieszka Zapora-Kurel
- 2nd Department of Nephrology with Hypertension and Dialysis, Medical University of Bialystok, 15-089 Bialystok, Poland; (A.Z.-K.); (M.Z.)
| | - Łukasz Kuźma
- Department of Invasive Cardiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (L.K.); (S.D.)
| | - Magdalena Zakrzewska
- 2nd Department of Nephrology with Hypertension and Dialysis, Medical University of Bialystok, 15-089 Bialystok, Poland; (A.Z.-K.); (M.Z.)
| | - Marcin Żórawski
- Department of Clinical Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland;
| | - Sławomir Dobrzycki
- Department of Invasive Cardiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (L.K.); (S.D.)
| | | | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-599-2660
| |
Collapse
|
17
|
Xia Y, Li Y, Wu X, Zhang Q, Chen S, Ma X, Yu M. Ironing Out the Details: How Iron Orchestrates Macrophage Polarization. Front Immunol 2021; 12:669566. [PMID: 34054839 PMCID: PMC8149954 DOI: 10.3389/fimmu.2021.669566] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Iron fine-tunes innate immune responses, including macrophage inflammation. In this review, we summarize the current understanding about the iron in dictating macrophage polarization. Mechanistically, iron orchestrates macrophage polarization through several aspects, including cellular signaling, cellular metabolism, and epigenetic regulation. Therefore, iron modulates the development and progression of multiple macrophage-associated diseases, such as cancer, atherosclerosis, and liver diseases. Collectively, this review highlights the crucial role of iron for macrophage polarization, and indicates the potential application of iron supplementation as an adjuvant therapy in different inflammatory disorders relative to the balance of macrophage polarization.
Collapse
Affiliation(s)
- Yaoyao Xia
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yikun Li
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoyan Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingzhuo Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siyuan Chen
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xianyong Ma
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Miao Yu
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
18
|
Filimon MN, Dumitrescu G, Caraba IV, Sinitean A, Verdes D, Mituletu M, Cornianu M, Popescu R. Effects of mine waste water on rat: bioaccumulation and histopathological evaluation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:20222-20239. [PMID: 33410069 DOI: 10.1007/s11356-020-11844-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/25/2020] [Indexed: 06/12/2023]
Abstract
The highlighting of the bioaccumulation capacity of metals in the internal organs, the mode of distribution at the level of internal organs, the interactions between them, respectively, and the histological changes occurred at the level of the liver and kidneys are the main aspects addressed in the present study. The experiment was performed on 4 groups of Wistar rats: 3 groups which were administered water from rivers located in the vicinity of the Bor mining operation and 1 control group. The determination of the metal content in the administered water samples and in the internal organs was performed using the flame atomic absorption spectrophotometer. Tissue alterations were assessed by histological technique and hematoxylin-eosin staining. The metal retention capacity in the internal organs differs depending on the metal concentration in the administered water sample but also on the organ in which the determination was made. Also, correlations were established between the concentrations of metals at the level of the organs, showing (a) positive and significant correlations-at the level of the heart between Zn and Cu, Fe, and Mn and at the level of the lungs between Mn and Cd-but the most numerous were reported in the testicle; (b) moderate correlations at liver level between Fe and Zn, at spleen level between Cu and Mn and Cd and at the level of the kidneys between Pb and Zn, Cu, and Fe; (c) negative correlations at renal level between Pb and Mn; and (d) insignificant correlations between Pb and Fe. The histological changes identified at the level of the liver and kidney become more obvious, and their aggravation is registered with the increase of the metal content.
Collapse
Affiliation(s)
- Marioara Nicoleta Filimon
- Department Biology-Chemistry, Faculty Chemistry-Biology-Geography, West University of Timisoara, Pestalozzi 16, RO 300315, Timisoara, Romania
| | - Gabi Dumitrescu
- Faculty of Bioengineering of Animal Resources, Banat University of Agricultural Sciences and Veterinary Medicine "King Mihai I of Romania" from Timisoara, Calea Aradului 119, RO 300645, Timisoara, Romania.
| | - Ion Valeriu Caraba
- Faculty of Bioengineering of Animal Resources, Banat University of Agricultural Sciences and Veterinary Medicine "King Mihai I of Romania" from Timisoara, Calea Aradului 119, RO 300645, Timisoara, Romania
| | - Adrian Sinitean
- Department Biology-Chemistry, Faculty Chemistry-Biology-Geography, West University of Timisoara, Pestalozzi 16, RO 300315, Timisoara, Romania
| | - Doina Verdes
- Department of Morphologic microscopy, University of Medicine and Pharmacy "Victor Babes", E. Murgu 2, RO 300041, Timisoara, Romania
| | - Mihai Mituletu
- Department of Morphologic microscopy, University of Medicine and Pharmacy "Victor Babes", E. Murgu 2, RO 300041, Timisoara, Romania
| | - Marioara Cornianu
- Department of Morphologic microscopy, University of Medicine and Pharmacy "Victor Babes", E. Murgu 2, RO 300041, Timisoara, Romania
| | - Roxana Popescu
- Department of Morphologic microscopy, University of Medicine and Pharmacy "Victor Babes", E. Murgu 2, RO 300041, Timisoara, Romania
| |
Collapse
|
19
|
McCullough PA. Anemia of cardiorenal syndrome. Kidney Int Suppl (2011) 2021; 11:35-45. [PMID: 33777494 PMCID: PMC7983020 DOI: 10.1016/j.kisu.2020.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 01/28/2023] Open
Abstract
Cardiorenal syndrome includes a spectrum of disorders of the kidneys and heart in which loss of function in one organ contributes to reduced function in the other organ. Cardiorenal syndrome is frequently complicated by comorbid anemia, which leads to reciprocal and progressive cardiac and renal deterioration. The triad of heart failure, chronic kidney disease (CKD), and anemia is termed cardiorenal anemia syndrome (CRAS). There are currently no evidence-based recommendations for managing patients with CRAS; however, the treatment of these patients is multifactorial. Not only must the anemia be controlled, but heart failure and kidney injury must be addressed, in addition to other comorbidities. Intravenous iron and erythropoiesis-stimulating agents are the mainstays of treatment for anemia of CKD, addressing both iron and erythropoiesis deficiencies. Since erythropoiesis-stimulating agent therapy can be associated with adverse outcomes at higher doses in patients with CKD and is not used in routine practice in patients with heart failure, treatment options for managing anemia in patients with CRAS are limited. Several new therapies, particularly the hypoxia-inducible factor-prolyl hydroxylase inhibitors, are currently under clinical development. The hypoxia-inducible factor-prolyl hydroxylase inhibitors have shown promising results for treating anemia of CKD in clinical trials and may confer benefits in patients with CRAS, potentially addressing some of the limitations of erythropoiesis-stimulating agents. Updated clinical practice guidelines for the screening and management of anemia in cardiorenal syndrome, in light of potential new therapies and clinical evidence, would improve the clinical outcomes of patients with this complex syndrome.
Collapse
Affiliation(s)
- Peter A. McCullough
- Department of Medicine, Texas A & M College of Medicine, Baylor University Medical Center, Baylor Heart and Vascular Hospital, Baylor Heart and Vascular Institute, Dallas, Texas, USA
| |
Collapse
|
20
|
Simultaneous management of disordered phosphate and iron homeostasis to correct fibroblast growth factor 23 and associated outcomes in chronic kidney disease. Curr Opin Nephrol Hypertens 2021; 29:359-366. [PMID: 32452919 DOI: 10.1097/mnh.0000000000000614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Hyperphosphatemia, iron deficiency, and anemia are powerful stimuli of fibroblast growth factor 23 (FGF23) production and are highly prevalent complications of chronic kidney disease (CKD). In this manuscript, we put in perspective the newest insights on FGF23 regulation by iron and phosphate and their effects on CKD progression and associated outcomes. We especially focus on new studies aiming to reduce FGF23 levels, and we present new data that suggest major benefits of combined corrections of iron, phosphate, and FGF23 in CKD. RECENT FINDINGS New studies show that simultaneously correcting iron deficiency and hyperphosphatemia in CKD reduces the magnitude of FGF23 increase. Promising therapies using iron-based phosphate binders in CKD might mitigate cardiac and renal injury and improve survival. SUMMARY New strategies to lower FGF23 have emerged, and we discuss their benefits and risks in the context of CKD. Novel clinical and preclinical studies highlight the effects of phosphate restriction and iron repletion on FGF23 regulation.
Collapse
|
21
|
A real-world longitudinal study of anemia management in non-dialysis-dependent chronic kidney disease patients: a multinational analysis of CKDopps. Sci Rep 2021; 11:1784. [PMID: 33469061 PMCID: PMC7815803 DOI: 10.1038/s41598-020-79254-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Previously lacking in the literature, we describe longitudinal patterns of anemia prescriptions for non-dialysis-dependent chronic kidney disease (NDD-CKD) patients under nephrologist care. We analyzed data from 2818 Stage 3-5 NDD-CKD patients from Brazil, Germany, and the US, naïve to anemia medications (oral iron, intravenous [IV] iron, or erythropoiesis stimulating agent [ESA]) at enrollment in the CKDopps. We report the cumulative incidence function (CIF) of medication initiation stratified by baseline characteristics. Even in patients with hemoglobin (Hb) < 10 g/dL, the CIF at 12 months for any anemia medication was 40%, and 28% for ESAs. Patients with TSAT < 20% had a CIF of 26% and 6% for oral and IV iron, respectively. Heart failure was associated with earlier initiation of anemia medications. IV iron was prescribed to < 10% of patients with iron deficiency. Only 40% of patients with Hb < 10 g/dL received any anemia medication within a year. Discontinuation of anemia treatment was very common. Anemia treatment is initiated in a limited number of NDD-CKD patients, even in those with guideline-based indications to treat. Hemoglobin trajectory and a history of heart failure appear to guide treatment start. These results support the concept that anemia is sub-optimally managed among NDD-CKD patients in the real-world setting.
Collapse
|
22
|
Zheng Q, Wang Y, Yang H, Sun L, Fu X, Wei R, Liu YN, Liu WJ. Efficacy and Safety of Daprodustat for Anemia Therapy in Chronic Kidney Disease Patients: A Systematic Review and Meta-Analysis. Front Pharmacol 2021; 11:573645. [PMID: 33597868 PMCID: PMC7883598 DOI: 10.3389/fphar.2020.573645] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Objective: Daprodustat is a novel oral agent in treating anemia of chronic kidney disease (CKD), and several clinical trials have been conducted to compare daprodustat with recombinant human erythropoietin (rhEPO) or placebo. Our systematic review aimed to investigate the efficacy and safety of daprodustat for anemia treatment in both dialysis-dependent (DD) and non-dialysis-dependent (NDD) patients. Methods: Six databases were searched for randomized controlled trials (RCTs) reporting daprodustat vs. rhEPO or placebo for anemia patients in CKD. The outcome indicators were focused on hemoglobin (Hb), ferritin, transferrin saturation (TSAT), total iron-binding capacity (TIBC), vascular endothelial growth factor (VEGF), and serious adverse events (SAEs). Results: Eight eligible studies with 1,516 participants were included. For both NDD and DD patients, changes in Hb levels from baseline were significantly higher in daprodustat group than that in the placebo (mean difference (MD) = 1.73, [95% confidence interval (CI), 0.34 to 3.12], p = 0.01; MD = 1.88, [95% CI, 0.68 to 3.09], p = 0.002; respectively), and there was no significant difference between daprodustat and rhEPO group (MD = 0.05, [95% CI, −0.49 to 0.59], p = 0.86; MD = 0.12, [95% CI, −0.28 to 0.52], p = 0.55; respectively). The indexes of iron metabolism were improved significantly in the daprodustat group compared to placebo- or rhEPO-treated patients, while there was no similar change in terms of TSAT for DD patients. Furthermore, no trend of increasing plasma VEGF was observed in daprodustat-treated subjects. As for safety, there was no significant difference in the incidence of SAEs between daprodustat and placebo treatment, while the incidence of SAEs in the daprodustat group was significantly lower than that in the rhEPO group. Conclusion: Daprodustat was efficacious and well tolerated for anemia in both NDD and DD patients in the short term based on current RCTs. And daprodustat may become an effective alternative for treatment of anemia with CKD. Since the application of daprodustat is still under exploration, future researches should consider the limitations of our study to evaluate the value of daprodustat.
Collapse
Affiliation(s)
- Qiyan Zheng
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yahui Wang
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Huisheng Yang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luying Sun
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xinwen Fu
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ruojun Wei
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yu Ning Liu
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Institute of Nephrology, and Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
23
|
Alageeli AA, Alqahtany FS, Algahtani FH. The Role of Reticulocyte Hemoglobin Content for the Diagnosis of Functional Iron Deficiency in Hemodialyzed patients. Saudi J Biol Sci 2021; 28:50-54. [PMID: 33424282 PMCID: PMC7783634 DOI: 10.1016/j.sjbs.2020.08.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 11/26/2022] Open
Abstract
The effectiveness of reticulocyte hemoglobin content (CHr) had been reported to detect early functional iron deficiency especially among Chronic kidney disease (CKD) patients. CHr is more superior to classic biochemical indices in reflecting transient iron-deficiency status, therefore improving diagnosis and treatment. This study was conducted to determine the sensitivity of CHr in the diagnosis of functional iron deficiency (FID) in hemodialyzed patients. One hundred hemodialyzed patients along with 60 healthy controls were recruited and blood specimens were collected. Venous blood was used for hematological and biochemical investigations collected via 3 ml lavender-top tubes for hematological tests including CBC, blood film, ESR and CHr, and red-top tube for biochemical tests including TIBC, SF and CRP. A statistically significant decrease was noted in CHr values between hemodialysis patients and the control group (24.8 ± 2.0 pg vs. 30.9 ± 1.3 pg, p<0.001). CHr values showed a significant correlations with RBCs, Hb- hemoglobin, Hct- hematocrit level, MCV- mean corpuscular volume, MCH- mean corpuscular hemoglobin, MCHC, RDW- red cell distribution width , SI-Serum Iron, TIBC- Total iron binding capacity and TSAT- Transferrin saturation. The present study showed that CHr in comparison to the conventional hematological and biochemical markers commonly used to diagnose iron deficiency.
Collapse
Affiliation(s)
- Ali A. Alageeli
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Fatmah S. Alqahtany
- Department of Pathology, Hematopathology Unit, College of Medicine, King Saud University, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Farjah H. Algahtani
- Department of Medicine, Division of Oncology/Hematology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Maxwell KD, Chuang J, Chaudhry M, Nie Y, Bai F, Sodhi K, Liu J, Shapiro JI. The potential role of Na-K-ATPase and its signaling in the development of anemia in chronic kidney disease. Am J Physiol Renal Physiol 2020; 320:F234-F242. [PMID: 33356956 DOI: 10.1152/ajprenal.00244.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the most prominent diseases affecting our population today. According to the Factsheet published by Centers for Disease Control and Prevention (CDC), it effects approximately 15% of the total population in the United States in some way, shape, or form. Within the myriad of symptomatology associated with CKD, one of the most prevalent factors in terms of affecting quality of life is anemia. Anemia of CKD cannot be completely attributed to one mechanism or cause, but rather has a multifactorial origin in the pathophysiology of CKD. While briefly summarizing well-documented risk factors, this review, as a hypothesis, aims to explore the possible role of Na-K-ATPase and its signaling function [especially recent identified reactive oxygen species (ROS) amplification function] in the interwoven mechanisms of development of the anemia of CKD.
Collapse
Affiliation(s)
- Kyle D Maxwell
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Justin Chuang
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Muhammad Chaudhry
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ying Nie
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Fang Bai
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Komal Sodhi
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia.,Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Jiang Liu
- Department of Biomsedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
25
|
Wojtaszek E, Glogowski T, Malyszko J. Iron and Chronic Kidney Disease: Still a Challenge. Front Med (Lausanne) 2020; 7:565135. [PMID: 33392212 PMCID: PMC7775475 DOI: 10.3389/fmed.2020.565135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022] Open
Abstract
Anemia is a clinical feature of chronic kidney disease (CKD). Most common causes are iron and erythropoietin deficiency. The last two decades have yielded significant advances in understanding iron balance's physiology, including iron trafficking and the crosstalk between iron, oxygen, and erythropoiesis. This knowledge sheds new light on the regulation and disturbance of iron homeostasis in CKD and holds the promise for developing new diagnostic and therapeutic tools to improve the management of iron disorders. Hepcidin-ferroportin axis has a central role in regulating body iron balance and coordinating communication between tissues and cells that acquire, store, and utilize iron. Recent research has revealed a bidirectional relationship between fibroblast growth factor 23 (FGF23) and iron status, anemia, and inflammation, as well as the role of erythroferrone (ERFE) in iron homeostasis. However, ERFE concentrations and actions are not well-characterized in CKD patients. Studies on ERFE in CKD are limited with slightly conflicting results. Despite general interest in iron metabolism in kidney diseases, studies on the less prevalent renal replacement therapy mode, such as peritoneal dialysis or hemodiafiltration, are scarce. Slightly more was published on hemodialysis. There are several novel options on the horizon; however, clinical data are limited. One should be aware of the potential risks and benefits of the novel, sophisticated therapies. An inhibition of hepcidin on the different pathways might be also a viable adjunctive therapeutic option in other clinical situations.
Collapse
Affiliation(s)
| | | | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Diseases, The Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
26
|
Plasma Protein Carbonyls as Biomarkers of Oxidative Stress in Chronic Kidney Disease, Dialysis, and Transplantation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2975256. [PMID: 33299524 PMCID: PMC7707964 DOI: 10.1155/2020/2975256] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/03/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
Accumulating evidence indicates that oxidative stress plays a role in the pathophysiology of chronic kidney disease (CKD) and its progression; during renal replacement therapy, oxidative stress-derived oxidative damage also contributes to the development of CKD systemic complications, such as cardiovascular disease, hypertension, atherosclerosis, inflammation, anaemia, and impaired host defence. The main mechanism underlying these events is the retention of uremic toxins, which act as a substrate for oxidative processes and elicit the activation of inflammatory pathways targeting endothelial and immune cells. Due to the growing worldwide spread of CKD, there is an overwhelming need to find oxidative damage biomarkers that are easy to measure in biological fluids of subjects with CKD and patients undergoing renal replacement therapy (haemodialysis, peritoneal dialysis, and kidney transplantation), in order to overcome limitations of invasive monitoring of CKD progression. Several studies investigated biomarkers of protein oxidative damage in CKD, including plasma protein carbonyls (PCO), the most frequently used biomarker of protein damage. This review provides an up-to-date overview on advances concerning the correlation between plasma protein carbonylation in CKD progression (from stage 1 to stage 5) and the possibility that haemodialysis, peritoneal dialysis, and kidney transplantation improve plasma PCO levels. Despite the fact that the role of plasma PCO in CKD is often underestimated in clinical practice, emerging evidence highlights that plasma PCO can serve as good biomarkers of oxidative stress in CKD and substitutive therapies. Whether plasma PCO levels merely serve as biomarkers of CKD-related oxidative stress or whether they are associated with the pathogenesis of CKD complications deserves further evaluation.
Collapse
|
27
|
Jankowska EA, Tkaczyszyn M, Ponikowski P. Myocardial iron content in non-ischaemic cardiomyopathy: how much is known? Eur J Heart Fail 2020; 22:2047-2048. [PMID: 33034152 DOI: 10.1002/ejhf.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ewa A Jankowska
- Department of Heart Diseases, Wroclaw Medical University, and Centre for Heart Diseases, University Hospital, Wroclaw, Poland
| | - Michał Tkaczyszyn
- Department of Heart Diseases, Wroclaw Medical University, and Centre for Heart Diseases, University Hospital, Wroclaw, Poland
| | - Piotr Ponikowski
- Department of Heart Diseases, Wroclaw Medical University, and Centre for Heart Diseases, University Hospital, Wroclaw, Poland
| |
Collapse
|
28
|
Akizawa T, Nangaku M, Yonekawa T, Okuda N, Kawamatsu S, Onoue T, Endo Y, Hara K, Cobitz AR. Efficacy and Safety of Daprodustat Compared with Darbepoetin Alfa in Japanese Hemodialysis Patients with Anemia: A Randomized, Double-Blind, Phase 3 Trial. Clin J Am Soc Nephrol 2020; 15:1155-1165. [PMID: 32723804 PMCID: PMC7409739 DOI: 10.2215/cjn.16011219] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/02/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Daprodustat is an oral hypoxia-inducible factor prolyl hydroxylase inhibitor that stimulates erythropoiesis and regulates genes related to iron metabolism. The efficacy (noninferiority) and safety of daprodustat compared with standard therapy (darbepoetin alfa) was evaluated. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This was a randomized, phase 3, double-blind, active-control study in Japanese patients receiving hemodialysis with anemia of CKD. Participants' treatment was switched from current erythropoiesis-stimulating agents (ESAs) to daprodustat 4 mg once daily or darbepoetin alfa 10-60 μg once weekly (on the basis of the prestudy ESA dose). Dose was adjusted every 4 weeks for daprodustat or every 2 weeks for darbepoetin alfa, according to a protocol-specified algorithm. The primary end point was mean hemoglobin during weeks 40-52 in the intent-to-treat population. RESULTS Of 332 participants screened, 271 participants were randomized (safety evaluation: 271 participants; efficacy evaluation: 267 intent-to-treat population). The mean hemoglobin during weeks 40-52 were maintained within the target range in both groups (10.9 g/dl [95% confidence interval (95% CI), 10.8 to 11.0] for daprodustat, and 10.8 g/dl [95% CI, 10.7 to 11.0] for darbepoetin alfa). Daprodustat was noninferior to darbepoetin alfa, as the lower bound of the confidence interval for the treatment difference (0.1 g/dl; 95% CI, -0.1 to 0.2 g/dl) was greater than the noninferiority criterion of -1.0 g/dl. For most participants, hemoglobin was maintained within the target range (10.0-12.0 g/dl) during weeks 40-52 (88% daprodustat; 90% darbepoetin alfa). Geometric mean hepcidin levels decreased more at week 52 with daprodustat (-37%; 95% CI, -49 to -23) than with darbepoetin alfa (-20%; 95% CI, -36 to -1), and an increase in total iron-binding capacity was observed in the daprodustat group. Frequency of adverse events were generally similar between daprodustat and darbepoetin alfa. CONCLUSIONS Oral daprodustat was noninferior to darbepoetin alfa as measured by mean hemoglobin over weeks 40-52 in Japanese patients receiving hemodialysis switched from ESAs. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER 201754, Clinicaltrials.gov, NCT02969655.
Collapse
Affiliation(s)
- Tadao Akizawa
- Division of Nephrology, Showa University School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taeko Yonekawa
- Medicines Development, Japan Development, GlaxoSmithKline, Tokyo, Japan
| | - Nobuhiko Okuda
- Medicines Development, Japan Development, GlaxoSmithKline, Tokyo, Japan
| | - Shinya Kawamatsu
- Clinical Operations, Japan Development, GlaxoSmithKline, Tokyo, Japan
| | - Tomohiro Onoue
- Biomedical Data Sciences, Japan Development, GlaxoSmithKline, Tokyo, Japan
| | - Yukihiro Endo
- Medicines Development, Japan Development, GlaxoSmithKline, Tokyo, Japan
| | - Katsutoshi Hara
- Clinical Pharmacology, Japan Development, GlaxoSmithKline, Tokyo, Japan
| | | |
Collapse
|
29
|
Liposomal iron in moderate chronic kidney disease. Nefrologia 2020; 40:446-452. [PMID: 31892487 DOI: 10.1016/j.nefro.2019.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/21/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION AND OBJECTIVE The optimal iron supplementation route of administration (intravenous vs oral) in patients with chronic kidney disease (CKD) not on dialysis is a hot topic of debate. An oral preparation (liposomal iron, FeSu) has recently been developed with high bioavailability and low incidence of side effects. The objective was to evaluate the efficacy of FeSu in patients with stage 3 CKD and gastrointestinal intolerance to conventional oral iron therapy. MATERIAL AND METHODS Prospective observational study of patients with stable stage 3 CKD and gastrointestinal intolerance to conventional oral iron therapy. An oral 30mg/day dose of FeSu was administered for 12 months. The primary outcome measure was haemoglobin increase at 6 and 12 months. Treatment adherence and adverse effects were also evaluated. RESULTS 37 patients aged 72.6±14.7 years and with an estimated glomerular filtration rate (eGFR) of 42±10ml/min/1.73m2 were included. 32 patients had received previous treatment with conventional oral formulations, 73% of which exhibited gastrointestinal intolerance with treatment adherence of 9.4%. After 6 months with FeSu, an increase in haemoglobin was observed versus baseline, which was sustained at 12 months (0.49±0.19 and 0.36±0.19g/dl, respectively, P<.05), despite a significant eGFR decrease of 3.16±1.16 and 4.20±1.28ml/min/1.73 m2 at 6 and 12 months, respectively. None of the patients experienced adverse reactions that required the treatment to be suspended. Adherence was 100% at both 6 and 12 months. CONCLUSIONS FeSu is effective in a cohort of patients with stage 3 CKD with similar characteristics to the general population of moderate CKD patients, with a low rate of adverse reactions and excellent tolerability.
Collapse
|
30
|
Souza E, Cho KH, Harris ST, Flindt NR, Watt RK, Pai AB. Hypoxia-inducible factor prolyl hydroxylase inhibitors: a paradigm shift for treatment of anemia in chronic kidney disease? Expert Opin Investig Drugs 2020; 29:831-844. [PMID: 32476498 DOI: 10.1080/13543784.2020.1777276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The hypoxia-inducible factor prolyl hydroxylase (HIF-PH) pathway is responsible for regulating the biosynthesis of erythropoietin (EPO) and maintaining iron homeostasis. Investigational drugs that target the HIF-PH pathway are promising alternatives for treating anemia in Chronic Kidney Disease (CKD). AREAS COVERED This review summarizes recent advances focused on the clinical development of HIF-PH inhibitors (HIF-PHIs) as potentially novel therapies in the treatment of anemia in CKD based on publications available on PubMed and restricted Google searches. We provide a comparison between HIF-PHIs regarding their pharmacokinetics, dosing regimens and safety concerns, structure-activity relationships, and alterations in key laboratory parameters observed in animal models and clinical trials. EXPERT OPINION HIF-PHIs may be advantageous in some aspects compared to the conventional erythropoiesis-stimulating agents (ESAs). While ESAs could increase the risk of cardiovascular events due to rapid rises in ESA blood levels, HIF-PHIs have been reported to maintain EPO concentrations at levels that are closer to the normal physiological ranges. Although HIF-PHIs have been demonstrated to be relatively safe and effective in clinical trials, long-term safety data are needed in order to establish whether these therapeutic agents will lead to a major paradigm change in the treatment of anemia of CKD.
Collapse
Affiliation(s)
- Ernane Souza
- Department of Clinical Pharmacy, University of Michigan , Ann Arbor, MI, USA
| | - Katherine H Cho
- Department of Clinical Pharmacy, University of Michigan , Ann Arbor, MI, USA
| | - Shelby T Harris
- Department of Chemistry and Biochemistry, Brigham Young University , Provo, UT, USA
| | - Naomi R Flindt
- Department of Chemistry and Biochemistry, Brigham Young University , Provo, UT, USA
| | - Richard K Watt
- Department of Chemistry and Biochemistry, Brigham Young University , Provo, UT, USA
| | - Amy Barton Pai
- Department of Clinical Pharmacy, University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
31
|
Fakharzadeh S, Argani H, Torbati PM, Dadashzadeh S, Kalanaky S, Nazaran MH, Basiri A. DIBc nano metal-organic framework improves biochemical and pathological parameters of experimental chronic kidney disease. J Trace Elem Med Biol 2020; 61:126547. [PMID: 32460199 DOI: 10.1016/j.jtemb.2020.126547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The growing morbidity and mortality rate of chronic kidney disease (CKD) has forced researchers to find more efficient strategies for controlling this disease. Studies have proven the important role of alteration in iron, zinc and selenium metabolism in CKD pathological process. Nanotechnology, through synthetizing nano metal-organic framework (NMOF) structures, can be employed as a valuable strategy for using these trace elements as the key for modification and improvement of CKD-related pathological events. After proving the anti-diabetic property of DIBc NMOF (which contains selenium and zinc) in the previous study, the impact of this NMOF on some important biochemical and pathological parameters of CKD was evaluated in the current study. METHODS Knowing that diabetic nephropathy (DN) is the leading cause of CKD, male wistar rats were selected and given a high fat diet for 2 weeks and then were injected with streptozotocin (35 mg/kg) to induce DN. Six weeks after streptozotocin injection, DIBc or metformin treatment started and continued for 8 weeks. RESULTS Eight weeks of DIBc treatment decreased plasma fasting blood glucose, blood urea nitrogen, uric acid, malondialdehyde (MDA) and HOMA-IR index compared to DN control and metformin groups. This NMOF significantly reduced urinary albumin excretion rate, MDA and 8-isoprostane, while it increased creatinine clearance in comparison to the above-mentioned groups. Renal histo-pathological images indicated that DIBc ameliorated glomerular basement membrane thickening and wrinkling, mesangial matrix expansion and hypercellularity and presence of intra-cytoplasmic hyaline droplets in proximal cortical tubules of kidney samples. CONCLUSION The results showed the therapeutic effect of DIBc on important biochemical and histo-pathological parameters of CKD, so this NMOF could be regarded as a promising novel anti-CKD agent.
Collapse
Affiliation(s)
- Saideh Fakharzadeh
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Hassan Argani
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Mohammadi Torbati
- Department of Pathology, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Kalanaky
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | | | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Abstract
Ferritins are evolutionarily conserved proteins that regulate cellular iron metabolism. It is the only intracellular protein that is capable of storing large quantities of iron. Although the ratio of different subunits determines the iron content of each ferritin molecule, the exact mechanism that dictates organization of these subunits still is unclear. In this review, we address renal ferritin expression and its implication in kidney disease. Specifically, we address the role of ferritin subunits in preventing kidney injury and also promoting tolerance against infection-associated kidney injury. We describe functions for ferritin that are independent of its ability to ferroxidize and store iron. We further discuss the implications of ferritin in body fluids, including blood and urine, during inflammation and kidney disease. Although there are several in-depth review articles on ferritin in the context of iron metabolism, we chose to focus on the role of ferritin particularly in kidney health and disease and highlight unanswered questions in the field.
Collapse
Affiliation(s)
- Kayla McCullough
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Subhashini Bolisetty
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
33
|
Bailey CK, Caltabiano S, Cobitz AR, Huang C, Mahar KM, Patel VV. A randomized, 29-day, dose-ranging, efficacy and safety study of daprodustat, administered three times weekly in patients with anemia on hemodialysis. BMC Nephrol 2019; 20:372. [PMID: 31619187 PMCID: PMC6796426 DOI: 10.1186/s12882-019-1547-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/29/2019] [Indexed: 12/27/2022] Open
Abstract
Background Daprodustat is a hypoxia-inducible factor-prolyl hydroxylase inhibitor currently being investigated as a treatment for anemia of chronic kidney disease (CKD) in both dialysis and nondialysis patients. In clinical studies to date, daprodustat has been administered orally as a once-daily regimen. This randomized, double-blind, placebo-controlled study characterized the initial dose-hemoglobin response as well as the efficacy and safety of three times weekly (TIW) daprodustat in hemodialysis patients switched from stable recombinant human erythropoietin (rhEPO), in accordance with a TIW hemodialysis schedule. Methods 103 patients on hemodialysis with baseline hemoglobin of 9.0 to 11.5 g/dL and previously receiving a stable dose of rhEPO or its analogs were randomized 1:1:1:1:1 to receive daprodustat 10, 15, 25, or 30 mg or placebo TIW over 29 days. Results Mean baseline hemoglobin was 10.6 g/dL for the placebo group and each daprodustat cohort. Daprodustat produced dose-dependent changes in mean hemoglobin from baseline to day 29. Using a Bayesian approach, the estimated dose conversion ratio between once-daily and TIW daprodustat was ~ 2.0 across the evaluated dose range using an Emax model. Daprodustat was generally well tolerated, with an adverse event (AE) profile consistent with the hemodialysis population. Conclusions These data help inform the appropriate dose conversion ratio to be applied to daily doses to obtain equivalent daprodustat TIW doses and suggest TIW treatment with daprodustat can treat anemia of CKD safely, supporting future long-term studies for this indication using a TIW dosing regimen. Trial registration ClinicalTrials.gov Identifier: NCT02689206; date registered: 02/11/2016. Electronic supplementary material The online version of this article (10.1186/s12882-019-1547-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine K Bailey
- R&D, Clinical Sciences, GlaxoSmithKline, 1250 S. Collegeville Road, Mail Code UP 4200, Collegeville, PA, 19426, USA.
| | - Stephen Caltabiano
- R&D, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Alexander R Cobitz
- R&D, Clinical Sciences, GlaxoSmithKline, 1250 S. Collegeville Road, Mail Code UP 4200, Collegeville, PA, 19426, USA
| | - Chun Huang
- R&D, Clinical Statistics, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Kelly M Mahar
- R&D, Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Vickas V Patel
- R&D, Discovery Medicine, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| |
Collapse
|
34
|
T2 ∗ Relaxation Time Obtained from Magnetic Resonance Imaging of the Liver Is a Useful Parameter for Use in the Construction of a Murine Model of Iron Overload. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:7463047. [PMID: 31598113 PMCID: PMC6778918 DOI: 10.1155/2019/7463047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 03/08/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022]
Abstract
Aim Iron overload is a life-threatening disorder that can increase the risks of cancer, cardiovascular disease, and liver cirrhosis. There is also a risk of iron overload in patients with chronic kidney disease. In patients with renal failure, iron storage is increased due to inadequate iron utilization associated with decreased erythropoiesis and also to the inflammatory status. To evade the risk of iron overload, an accurate and versatile indicator of body iron storage in patients with iron overload is needed. In this study, we aimed to find useful iron-related parameters that could accurately reflect body iron storage in mice in order to construct a murine model of iron overload. Methods To select an appropriate indicator of body iron status, a variety of parameters involved in iron metabolism were evaluated. Noninvasively measured parameters were R1, R2, and R2∗ derived from magnetic resonance imaging (MRI). Invasively measured parameters included serum hepcidin levels, serum ferritin levels, and liver iron contents. Histopathological analysis was also conducted. Results/Conclusion Among the several parameters evaluated, the MRI T2∗ relaxation time was able to detect iron storage in the liver as sensitively as serum ferritin levels. Moreover, it is expected that using an MRI parameter will allow accurate evaluation of body iron storage in mice over time.
Collapse
|
35
|
Anemia of Inflammation with An Emphasis on Chronic Kidney Disease. Nutrients 2019; 11:nu11102424. [PMID: 31614529 PMCID: PMC6835368 DOI: 10.3390/nu11102424] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 01/28/2023] Open
Abstract
Iron is vital for a vast variety of cellular processes and its homeostasis is strictly controlled and regulated. Nevertheless, disorders of iron metabolism are diverse and can be caused by insufficiency, overload or iron mal-distribution in tissues. Iron deficiency (ID) progresses to iron-deficiency anemia (IDA) after iron stores are depleted. Inflammation is of diverse etiology in anemia of chronic disease (ACD). It results in serum hypoferremia and tissue hyperferritinemia, which are caused by elevated serum hepcidin levels, and this underlies the onset of functional iron-deficiency anemia. Inflammation is also inhibitory to erythropoietin function and may directly increase hepcidin level, which influences iron metabolism. Consequently, immune responses orchestrate iron metabolism, aggravate iron sequestration and, ultimately, impair the processes of erythropoiesis. Hence, functional iron-deficiency anemia is a risk factor for several ailments, disorders and diseases. Therefore, therapeutic strategies depend on the symptoms, severity, comorbidities and the associated risk factors of anemia. Oral iron supplements can be employed to treat ID and mild anemia particularly, when gastrointestinal intolerance is minimal. Intravenous (IV) iron is the option in moderate and severe anemic conditions, for patients with compromised intestinal integrity, or when oral iron is refractory. Erythropoietin (EPO) is used to treat functional iron deficiency, and blood transfusion is restricted to refractory patients or in life-threatening emergency situations. Despite these interventions, many patients remain anemic and do not respond to conventional treatment approaches. However, various novel therapies are being developed to treat persistent anemia in patients.
Collapse
|
36
|
Hua YL, Ma Q, Yuan ZW, Zhang XS, Yao WL, Ji P, Hu JJ, Wei YM. A novel approach based on metabolomics coupled with network pharmacology to explain the effect mechanisms of Danggui Buxue Tang in anaemia. Chin J Nat Med 2019; 17:275-290. [PMID: 31076131 DOI: 10.1016/s1875-5364(19)30031-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Danggui Buxue Tang (DBT) is a famous Chinese medicinal decoction. Mechanism of DBT action is wide ranging and unclear. Exploring new ways of treatment with DBT is useful. Sprague-Dawley(SD) rats were randomly divided into 3 groups including control (NC, Saline), the DBT (at a dose of 8.10 g-1), and blood deficiency(BD) (Cyclophosphamide (APH)-andCyclophosphamide(CTX)-induced anaemia). A metabolomics approach using Liquid Chromatography-Quadrupole-Time-of-Flight/Mass Spectrometry (LC/Q-TOFMS) was developed to perform the plasma metabolic profiling analysis and differential metaboliteswerescreened according to the multivariate statistical analysiscomparing the NC and BD groups, andthe hub metabolites were outliers with high scores of the centrality indices. Anaemia disease-related protein target and compound of DBT databases were constructed. The TCMSP, ChemMapper and STITCH databases were used to predict the protein targets of DBT. Using the Cytoscape 3.2.1 to establish a phytochemical component-target protein interaction network and establish a component, protein and hub metabolite protein-protein interaction (PPI) network and merging the three PPI networks basing on BisoGenet. The gene enrichment analysis was used to analyse the relationship between proteins based on the relevant genetic similarity by ClueGO. The results shown DBT effectively treated anaemia in vivo. 11 metabolic pathways are involved in the therapeutic effect of DBT in vivo; S-adenosyl-l-methionine, glycine, l-cysteine, arachidonic acid (AA) and phosphatidylcholine(PC) were screened as hub metabolites in APH-and CTX-induced anaemia. A total of 288 targets were identified as major candidates for anaemia progression. The gene-set enrichment analysis revealed that the targets are involved in iron ion binding, haemopoiesis, reactive oxygen species production, inflammation and apoptosis. The results also showed that these targets were associated with iron ion binding, haemopoiesis, ROS production, apoptosis, inflammation and related signalling pathways. DBT can promote iron ion binding and haemopoiesis activities, restrain inflammation, production of reactive oxygen, block apoptosis, and contribute significantly to the DBT treat anaemia.
Collapse
Affiliation(s)
- Yong-Li Hua
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China.
| | - Qi Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| | - Zi-Wen Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| | - Xiao-Song Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| | - Wan-Ling Yao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| | - Peng Ji
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| | - Jun-Jie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| | - Yan-Ming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070,China
| |
Collapse
|
37
|
Akizawa T, Macdougall IC, Berns JS, Yamamoto H, Taguchi M, Iekushi K, Bernhardt T. Iron Regulation by Molidustat, a Daily Oral Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor, in Patients with Chronic Kidney Disease. Nephron Clin Pract 2019; 143:243-254. [PMID: 31387097 PMCID: PMC6979436 DOI: 10.1159/000502012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/09/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND/AIMS The current treatment for anemia associated with chronic kidney disease (CKD) includes the administration of erythropoiesis stimulating agents (ESAs) combined with iron supplementation. Molidustat, a hypoxia-inducible factor prolyl hydroxylase inhibitor, has potential to treat anemia associated with CKD through increased erythropoietin production and improved iron availability. Here, we report the effect of molidustat on iron metabolism. METHOD Parameters of iron metabolism were monitored in three 16-week, randomized, controlled, phase 2 studies assessing the safety and efficacy of molidustat in the treatment of anemia associated with CKD in different populations: treatment-naïve and previously ESA-treated patients not on dialysis, and previously ESA-treated patients on hemodialysis. Iron supplementation was left at the discretion of the investigator. RESULTS In treatment-naïve patients not on dialysis, transferrin saturation (TSAT), hepcidin, ferritin, and iron concentrations decreased with molidustat, whereas total iron binding capacity (TIBC) increased. Similar results were observed in previously ESA-treated patients not on dialysis, although changes in those parameters were larger in treatment-naïve than in previously ESA-treated patients. In previously ESA-treated patients receiving hemodialysis, hepcidin concentration and TIBC remained stable with molidustat, whereas TSAT and ferritin and iron concentrations increased. Generally, similar trends were observed in secondary analyses of subgroups of patients not receiving iron supplementation. CONCLUSIONS Molidustat is a potential alternative to standard treatment of anemia associated with CKD, with a different mechanism of action. In patients not receiving dialysis, molidustat increases iron availability. In patients receiving hemodialysis, further investigation is required to understand fully the mechanisms underlying iron mobilization associated with molidustat.
Collapse
Affiliation(s)
- Tadao Akizawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan,
| | - Iain C Macdougall
- Department of Renal Medicine, King's College Hospital, London, United Kingdom
| | - Jeffrey S Berns
- Perelman School of Medicine at the University of Pennsylvania, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hiroyasu Yamamoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
38
|
Kshirsagar AV, Li X. Long-Term Risks of Intravenous Iron in End-Stage Renal Disease Patients. Adv Chronic Kidney Dis 2019; 26:292-297. [PMID: 31477259 DOI: 10.1053/j.ackd.2019.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 01/07/2023]
Abstract
Patients with end-stage renal disease on dialysis commonly receive intravenous iron to treat anemia along with erythropoiesis-stimulating agents. While studies of intravenous iron have demonstrated efficacy in raising hemoglobin, the quantity of administered intravenous iron has raised concerns about iron overload leading to long-term toxicities. The goal of this review is to understand recent trends in intravenous iron use, potential mechanisms of iron toxicity, and to evaluate the available evidence in the literature for potential long-term cardiovascular and infectious complications. We include findings from the recently published landmark clinical trial of intravenous iron for patients receiving hemodialysis to contextualize treatment recommendations.
Collapse
|
39
|
Randomised clinical trial of ferric citrate hydrate on anaemia management in haemodialysis patients with hyperphosphataemia: ASTRIO study. Sci Rep 2019; 9:8877. [PMID: 31222044 PMCID: PMC6586649 DOI: 10.1038/s41598-019-45335-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023] Open
Abstract
Ferric citrate hydrate (FC) is an iron-based phosphate binder approved for hyperphosphataemia in patients with chronic kidney disease. We conducted a randomised controlled trial to evaluate the effects of FC on anaemia management in haemodialysis patients with hyperphosphataemia. We 1:1 randomised 93 patients who were undergoing haemodialysis and being treated with non-iron-based phosphate binders and erythropoiesis-stimulating agents (ESA) to receive 24 weeks of FC or to continue their non-iron-based phosphate binders (control) in a multicentre, open-label, parallel-design. Phosphate level was controlled within target range (3.5–6.0 mg/dL). The primary endpoint was change in ESA dose from baseline to end of treatment. Secondary endpoints were changes in red blood cell, iron and mineral, and bone-related parameters. Compared with control, FC reduced ESA dose [mean change (SD), −1211.8 (3609.5) versus +1195 (6662.8) IU/week; P = 0.03] without significant differences in haemoglobin. FC decreased red blood cell distribution width (RDW) compared with control. While there were no changes in serum phosphate, FC reduced C-terminal fibroblast growth factor (FGF) 23 compared with control. The incidence of adverse events did not differ significantly between groups. Despite unchanged phosphate and haemoglobin levels, FC reduced ESA dose, RDW, and C-terminal FGF23 compared with control.
Collapse
|
40
|
A Schiff Base Fluorescence Enhancement Probe for Fe(III) and Its Sensing Applications in Cancer Cells. SENSORS 2019; 19:s19112500. [PMID: 31159266 PMCID: PMC6603573 DOI: 10.3390/s19112500] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 12/15/2022]
Abstract
We report a new Schiff base fluorescent probe which senses ferric ion, Fe(III), with a significant fluorescence enhancement response. The probe showed high sensitivity (0.8 ppb), and fast response time (<10 s) of Fe(III) in aqueous media. In addition, the probe showed the ability to sense Fe(III) in a HeLa cancer cell line, with very low cytotoxicity. As a new bio-imaging probe for Fe(III), it gave bright fluorescent images in confocal laser scanning microscopy (CLSM).
Collapse
|
41
|
Serum Erythroferrone Levels Associate with Mortality and Cardiovascular Events in Hemodialysis and in CKD Patients: A Two Cohorts Study. J Clin Med 2019; 8:jcm8040523. [PMID: 30995819 PMCID: PMC6518296 DOI: 10.3390/jcm8040523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/05/2019] [Accepted: 04/05/2019] [Indexed: 02/07/2023] Open
Abstract
Erythroferrone (ERFE) is a hepcidin inhibitor whose synthesis is stimulated by erythropoietin, which increases iron absorption and mobilization. We studied the association between serum ERFE and mortality and non-fatal cardiovascular (CV) events in a cohort of 1123 hemodialysis patients and in a cohort of 745 stage 1–5 chronic kidney disease (CKD) patients. Erythroferrone was measured by a validated enzyme-linked immunosorbent assay (ELISA). In the hemodialysis cohort, serum ERFE associated directly with erythropoiesis stimulating agents (ESA) dose (p < 0.001) and inversely with serum iron and ferritin (p < 0.001). Erythroferrone associated with the combined outcome in an analysis adjusting for traditional risk factors, factors peculiar to end-stage kidney disease, serum ferritin, inflammation, and nutritional status (HR, hazard ratio, (5 ng/mL increase: 1.04, 95% confidence interval, CI: 1.01–1.08, p = 0.005). Furthermore, treatment with ESA modified the relationship between ERFE and the combined end-point in adjusted analyses (p for the effect modification = 0.018). Similarly, in CKD patients there was a linear increase in the risk for the same outcome in adjusted analyses (HR (2 ng/mL increase): 1.04, 95% CI: 1.0–1.07, p = 0.015). Serum ERFE is associated with mortality and CV events in CKD and in HD patients, and treatment by ESA amplifies the risk for this combined end-point in HD patients.
Collapse
|
42
|
Gołębiewska JE, Krawczyk B, Wysocka M, Ewiak A, Komarnicka J, Bronk M, Rutkowski B, Dębska-Ślizień A. Host and pathogen factors in Klebsiella pneumoniae upper urinary tract infections in renal transplant patients. J Med Microbiol 2019; 68:382-394. [PMID: 30747620 DOI: 10.1099/jmm.0.000942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To analyse the role of virulence factors (VFs) and host in Klebsiella pneumoniae upper urinary tract infections (UTIs) in renal transplant (RTx) recipients. METHODOLOGY Clinical and demographic data were registered prospectively. Phylogenetic background of K. pneumoniae isolates was analysed by PCR melting profiles (MP) and the following VFs genes: fimH-1, uge, kpn, ycfM, mrkD, rmpA, magA, hlyA, cnf-1, irp-1, irp-2, fyuA, entB, iutA, iroN by PCR. RESULTS We studied urine cultures and clinical data from 61 episodes of K. pneumoniae UTI in 54 RTx recipients. There were 32 cases of AB (53%), 10 cases of lower UTI (16%), 19 cases of AGPN (31%), including six cases of bacteraemia. In total, 74 % of strains were extended-spectrum beta-lactamase+, and there were two carbapenemase-producing strains. PCR MP typing showed a diverse population with 52 different genetic profiles of K. pneumoniae. Analysis of the DNA profiles indicated 45 unrelated, unique genotypes and 7 related (16 isolates from 15 patients) genotypes. Urine flow impairment emerged as an independent predictor of K. pneumoniae upper UTIs (OR 14.28, CI 2.7-75.56, P 0.002), while we did not find any association between the profile of VFs and developing upper UTIs. The prevalence of the uge gene was lower in RTx patients on everolimus when compared to isolates from patients not receiving mTOR inhibitors (33.3 % vs 82.8 % P<0.05). CONCLUSIONS K. pneumoniae upper UTI may be a marker of urine flow impairment. Bacterial VFs could not discriminate between upper and lower UTIs. However, immunosuppression may influence the selection of particular VFs.
Collapse
Affiliation(s)
- Justyna E Gołębiewska
- 1 Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, ul. Dębinki 7, 80-952 Gdańsk, Poland
| | - Beata Krawczyk
- 2 Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, ul. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Magdalena Wysocka
- 2 Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, ul. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Aleksandra Ewiak
- 3 Laboratory of Clinical Microbiology, University Centre for Laboratory Diagnostics, Medical University of Gdańsk Clinical Centre, ul. Dębinki 7, 80-952 Gdańsk, Poland
| | - Jolanta Komarnicka
- 3 Laboratory of Clinical Microbiology, University Centre for Laboratory Diagnostics, Medical University of Gdańsk Clinical Centre, ul. Dębinki 7, 80-952 Gdańsk, Poland
| | - Marek Bronk
- 3 Laboratory of Clinical Microbiology, University Centre for Laboratory Diagnostics, Medical University of Gdańsk Clinical Centre, ul. Dębinki 7, 80-952 Gdańsk, Poland
| | - Bolesław Rutkowski
- 1 Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, ul. Dębinki 7, 80-952 Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- 1 Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, ul. Dębinki 7, 80-952 Gdańsk, Poland
| |
Collapse
|
43
|
Hannedouche T, Fouque D, Joly D. Complications métaboliques en insuffisance rénale chronique : hyperphosphatémie, hyperkaliémie et anémie. Nephrol Ther 2018; 14:6S17-6S25. [DOI: 10.1016/s1769-7255(18)30647-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Kung WJ, Shih CT, Lee CH, Lin CC. The Divalent Elements Changes in Early Stages of Chronic Kidney Disease. Biol Trace Elem Res 2018; 185:30-35. [PMID: 29285723 DOI: 10.1007/s12011-017-1228-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/18/2017] [Indexed: 01/10/2023]
Abstract
As the glomerular filtration rate (GFR) decreases, it can cause imbalance in some divalent elements. These imbalances can cause increased oxidative stress in patients with renal impairment. The aim of present study was to investigate the changes of these divalent elements with CKD progression. One hundred and ninety-four patients with chronic kidney diseases (CKD) were divided into five stages, stage 1, 2, 3a, 3b, 4, and were recruited into this study. The divalent elements, calcium, magnesium, phosphorus, as well as iron, zinc, and copper were determined in clinical chemistry analyzer. Higher CKD stages were found to be associated with increased levels of phosphorus and copper; Ptrend values were 0.002 and 0.004, respectively. Also, higher CKD stages were associated with decreased levels of zinc; Ptrend value was 0.002, after adjustment for age, gender, smoke, education, diabetes, hypertension, and BMI. Decreased levels of zinc and elevated levels of phosphorus and copper might increase the oxidative stress and complications in CKD patients. Future randomized studies are needed to show whether adjusting dietary intake of phosphorus, copper, and zinc might affect the progression of CKD.
Collapse
Affiliation(s)
- Wan-Ju Kung
- Department of Laboratory Medicine, Fooyin University Hospital, Pingtung, Taiwan
| | - Ching-Tang Shih
- Department of Family Medicine, Fooyin University Hospital, Pingtung, Taiwan
| | - Chien-Hung Lee
- Department of Public Health and Environmental Medicine Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chiang Lin
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung, Taiwan.
- Department of Education and Research, Fooyin University Hospital, Pingtung, Taiwan.
| |
Collapse
|
45
|
McCullough PA, Uhlig K, Neylan JF, Pergola PE, Fishbane S. Usefulness of Oral Ferric Citrate in Patients With Iron-Deficiency Anemia and Chronic Kidney Disease With or Without Heart Failure. Am J Cardiol 2018; 122:683-688. [PMID: 29961562 DOI: 10.1016/j.amjcard.2018.04.062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 12/28/2022]
Abstract
Patients with chronic inflammatory conditions including chronic kidney disease (CKD) and heart failure (HF) are undertreated with iron-deficiency anemia (IDA). Progressive inflammation and reduced iron transport associated with CKD and HF may reduce the efficacy of oral iron therapy. Oral ferric citrate improves anemia markers in CKD, but its effects in patients with CKD and concomitant HF have not been described. Patients with CKD not on dialysis and IDA from a phase 2 and 3 trial were treated with ferric citrate (n = 190) or placebo (n = 188); patients with HF were identified from medical histories. Hemoglobin response was defined as a ≥10.0-g/L increase in hemoglobin. Changes in hemoglobin, transferrin saturation, ferritin, and serum phosphate from baseline to week 12 and the incidence of adverse events potentially related to HF were evaluated. HF was reported in 22% (n = 81) of patients. The proportion of patients with hemoglobin response to ferric citrate treatment did not significantly differ in patients with and without HF (43% vs 49%, respectively; p = 0.47); changes from baseline in hemoglobin, iron parameters, and serum phosphate were similar. Adverse events potentially related to HF were noted more frequently in patients with HF (ferric citrate, 23%; placebo, 17%) versus those without HF (ferric citrate, 12%; placebo, 11%). In conclusion, these results indicate a potential role for ferric citrate in the treatment of IDA in patients with CKD not on dialysis and concomitant HF.
Collapse
Affiliation(s)
- Peter A McCullough
- Baylor University Medical Center, Baylor Heart and Vascular Hospital, Baylor Heart and Vascular Institute, Dallas, Texas 75246.
| | - Katrin Uhlig
- Keryx Biopharmaceuticals, Inc., Boston, Massachusetts
| | - John F Neylan
- Keryx Biopharmaceuticals, Inc., Boston, Massachusetts
| | | | | |
Collapse
|
46
|
Rossaint J, Unruh M, Zarbock A. Fibroblast growth factor 23 actions in inflammation: a key factor in CKD outcomes. Nephrol Dial Transplant 2018; 32:1448-1453. [PMID: 27659127 DOI: 10.1093/ndt/gfw331] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
During chronic kidney disease (CKD), bone mineral metabolism is disturbed owing in part to the endogenous hormone fibroblast growth factor 23 (FGF23). Elevated FGF23 levels are seen in CKD patients. Current research has demonstrated that FGF23 directly modulates the immune response and host defense to bacterial infections. FGF23 also impairs the activation and recruitment of neutrophils, which are the main immune effector cells required for host defense against bacterial infections. In addition, while FGF23 levels reduce leukocyte recruitment and functions, inflammatory conditions may also-in a reverse fashion-contribute to elevated FGF23 levels in the circulation. In this context, altered hypoxia inducible factor 1α signaling and iron metabolism may contribute to intact FGF23 (iFGF23) production. This review examines evidence on the role of FGF23 in inflammation, immune cell function and recruitment as well as the regulation of FGF23 during inflammation and the clinical implications of this process for the immune system in individuals with CKD. Clinical observations and laboratory investigations indicate an important role of FGF23 in directly modulating leukocyte activation and recruitment behavior with consequences on host defense against bacterial infections. This novel observation may in part explain the increased infectious risk among patients with CKD. However, studies of FGF23 neutralization also revealed increased mortality after sustained administration over several weeks in rats. Thus, therapeutic interventions targeting FGF23 must be carefully evaluated.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Mark Unruh
- Division of Nephrology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA.,Section of Neprology, New Mexico Veteran Affairs Hospital, Albuquerque, NM, USA
| | - Alexander Zarbock
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
47
|
Wish JB, Aronoff GR, Bacon BR, Brugnara C, Eckardt KU, Ganz T, Macdougall IC, Núñez J, Perahia AJ, Wood JC. Positive Iron Balance in Chronic Kidney Disease: How Much is Too Much and How to Tell? Am J Nephrol 2018; 47:72-83. [PMID: 29439253 DOI: 10.1159/000486968] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Regulation of body iron occurs at cellular, tissue, and systemic levels. In healthy individuals, iron absorption and losses are minimal, creating a virtually closed system. In the setting of chronic kidney disease and hemodialysis (HD), increased iron losses, reduced iron absorption, and limited iron availability lead to iron deficiency. Intravenous (IV) iron therapy is frequently prescribed to replace lost iron, but determining an individual's iron balance and stores can be challenging and imprecise, contributing to uncertainty about the long-term safety of IV iron therapy. SUMMARY Patients on HD receiving judicious doses of IV iron are likely to be in a state of positive iron balance, yet this does not appear to confer an overt risk for clinically relevant iron toxicity. The concomitant use of iron with erythropoiesis-stimulating agents, the use of maintenance iron dosing regimens, and the reticuloendothelial distribution of hepatic iron deposition likely minimize the potential for iron toxicity in patients on HD. Key Messages: Because no single diagnostic test can, at present, accurately assess iron status and risk for toxicity, clinicians need to take an integrative approach to avoid iron doses that impose excessive exposure while ensuring sufficient replenishment of iron stores capable of overcoming hepcidin blockade and allowing for effective erythropoiesis.
Collapse
Affiliation(s)
- Jay B Wish
- Division of Nephrology, Indiana University Health, Indianapolis, Indiana, USA
| | - George R Aronoff
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
- DaVita Kidney Care, Denver, Colorado, USA
| | - Bruce R Bacon
- Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Carlo Brugnara
- Department of Laboratory Medicine, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin, Berlin, Germany
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Iain C Macdougall
- Department of Renal Medicine, King's College Hospital, Denmark Hill, London, United Kingdom
| | - Julio Núñez
- Cardiology Service, Hospital Clínico Universitario, INCLIVA, CIBERCV and University of Valencia, Valencia, Spain
| | - Adam J Perahia
- NorthStar Strategic Consulting, LLC, Gladstone, New Jersey, USA
| | - John C Wood
- Division of Cardiology, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor 23 (FGF23) is a hormone secreted by osteocytes and osteoblasts that regulates phosphorus and vitamin D homeostasis. FGF23 levels increase progressively in chronic kidney disease (CKD), and FGF23 excess might be a causal factor of left ventricular hypertrophy, CKD progression and death. Therefore, understanding the molecular mechanisms that control FGF23 production is critical to design therapies to lower FGF23 levels. The present review focuses on the role of inflammatory stimuli on FGF23 regulation and summarizes recent studies that support a novel framework linking inflammation to FGF23 regulation. RECENT FINDINGS Inflammation and iron deficiency, which are common occurrences in CKD, have emerged as novel FGF23 regulators. Recent findings show that inflammation increases FGF23 production in bone through direct and iron-related indirect mechanisms. In these settings, hypoxia-inducible factor (HIF)-1α orchestrates FGF23 transcription in response to inflammation and is primarily responsible for coordinating FGF23 production and cleavage. SUMMARY We demonstrate that inflammation increases FGF23 production and may contribute to elevated FGF23 levels in CKD. Osseous HIF-1α may represent a therapeutic target to lower FGF23 levels in CKD patients and minimize the negative consequences associated with FGF23 excess.
Collapse
|
49
|
Kawada S, Nagasawa Y, Kawabe M, Ohyama H, Kida A, Kato-Kogoe N, Nanami M, Hasuike Y, Kuragano T, Kishimoto H, Nakasho K, Nakanishi T. Iron-induced calcification in human aortic vascular smooth muscle cells through interleukin-24 (IL-24), with/without TNF-alpha. Sci Rep 2018; 8:658. [PMID: 29330517 PMCID: PMC5766506 DOI: 10.1038/s41598-017-19092-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/20/2017] [Indexed: 12/14/2022] Open
Abstract
In CKD patients, arteriosclerotic lesions, including calcification, can occur in vascular smooth muscle cells in a process called Moenckeberg's medial arteriosclerosis. Iron overload induces several complications, including the acceleration of arteriosclerosis. However, the relationship between Moenckeberg's arteriosclerosis in vascular smooth muscle cells and iron accumulation has remained unknown. We tested the accelerated effect of iron on calcification in cultured human aortic vascular smooth muscle cells (HASMCs). After establishment of this model, we performed a microarray analysis using mRNA from early stage culture HASMCs after iron stimulation with or without TNF-alpha stimulation. The role of interleukin-24 (IL-24) was confirmed from candidate genes that might contribute to calcification. HASMCs demonstrated calcification induced by iron and TNF-alpha. Calcification of HASMCs was synergistically enhanced by stimulation with both iron and TNF-alpha. In the early phase of calcification, microarray analysis revealed up-regulation of IL-24. Stimulation of HASMCs by IL-24 instead of iron induced calcification. The anti-IL-24 antibody reversed the effect of IL-24, supporting the important role of IL-24 in HASMCs calcification. In conclusion, iron-induced calcification in vascular smooth muscle cells occurred via IL-24, IL-24 was increased during the calcification process induced by iron, and IL-24 itself caused calcification in the absence of iron.
Collapse
Affiliation(s)
- Sayuri Kawada
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Yasuyuki Nagasawa
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan.
| | - Mutsuki Kawabe
- Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan.,Department of Oral and Maxillofacial Surgery, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Hideki Ohyama
- Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Aritoshi Kida
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Nahoko Kato-Kogoe
- Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Masayoshi Nanami
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Yukiko Hasuike
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Takahiro Kuragano
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Hiromitsu Kishimoto
- Department of Oral and Maxillofacial Surgery, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| | - Takeshi Nakanishi
- Department of Internal Medicine, Division of Kidney and Dialysis, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, Japan
| |
Collapse
|
50
|
Kim T, Streja E, Soohoo M, Rhee CM, Eriguchi R, Kim TW, Chang TI, Obi Y, Kovesdy CP, Kalantar-Zadeh K. Serum Ferritin Variations and Mortality in Incident Hemodialysis Patients. Am J Nephrol 2017; 46:120-130. [PMID: 28704813 DOI: 10.1159/000478735] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/09/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Higher serum ferritin levels may be influenced by iron use and inflammation, and are associated with higher mortality in hemodialysis (HD) patients. We hypothesized that a major rise in serum ferritin is associated with a higher risk of mortality, irrespective of baseline serum ferritin in incident HD patients. METHODS In a cohort of 93,979 incident HD patients between 2007 and 2011, we examined the association of change in serum ferritin from the baseline patient quarter (first 91 days from dialysis start) to the subsequent quarter with mortality. Multivariable adjustments were done for case-mix and markers of the malnutrition, and inflammation complex and intravenous iron dose. Change in serum ferritin was stratified into 5 groups: <-400, -400 to <-100, -100 to <100, 100 to <400, and ≥400 ng/mL/quarter. RESULTS The median change in serum ferritin was 89 ng/mL/quarter (interquartile range -55 to 266 ng/mL/quarter). Compared to stable serum ferritin (-100 to <100 ng/mL/quarter), a major rise (≥400 ng/mL/quarter) was associated with higher all-cause mortality (hazard ratio [95% CI] 1.07 [0.99-1.15], 1.17 [1.09-1.24], 1.26 [1.12-1.41], and 1.49 [1.27-1.76] according to baseline serum ferritin: <200, 200 to <500, 500 to <800, and ≥800 ng/mL in adjusted models, respectively. The mortality risk associated with a rise in serum ferritin was robust, irrespective of intravenous iron use. CONCLUSIONS During the first 6-months after HD initiation, a major rise in serum ferritin in those with a baseline ferritin ≥200 ng/mL and even a slight rise in serum ferritin in those with a baseline ferritin ≥800 ng/mL are associated with higher mortality.
Collapse
Affiliation(s)
- Taehee Kim
- Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine, School of Medicine, Orange, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|