1
|
Maier I, Kienzle E. A Meta-Analysis on Quantitative Calcium, Phosphorus and Magnesium Metabolism in Horses and Ponies. Animals (Basel) 2024; 14:2765. [PMID: 39409714 PMCID: PMC11475699 DOI: 10.3390/ani14192765] [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: 08/07/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The aims of the present meta-analysis were (i) to re-evaluate the factorially calculated Ca, P and Mg requirements to replace endogenous faecal losses, taking new data into account, (ii) to identify potential differences between horses and ponies regarding requirements, apparent digestibility, serum levels and renal excretion of Ca, P and Mg and (iii) to investigate the influence of mineral sources, i.e., "inorganic" sources from added mineral salts and "organic" sources from feed plants. For P, the water solubility of "inorganic" sources was taken into consideration. Data on the aforementioned parameters from 42 studies were plotted against intake, similar to the Lucas test for true digestibility and faecal endogenous losses. Within specific intake ranges, data were compared using t-tests and an ANOVA, followed by Holm-Sidak post hoc tests. Ponies had lower endogenous faecal Mg losses than horses. Consequently, apparent Mg digestibility was higher in ponies. Factorial calculations of Mg requirements to replace faecal losses showed that ponies needed approximately half of the current recommended amount, while horses required 1.9 times the amount currently recommended by Kienzle and Burger. The overall mean matched previous recommendations. For Ca, there was no discernible difference between ponies and horses. True Ca digestibility calculated by the Lucas test was higher and endogenous losses were lower when "organic" Ca was fed as opposed to when "inorganic" sources were used. The resulting factorial calculations of the requirements to replace faecal losses were close to current recommendations for "organic" Ca. For "inorganic" sources, however, the new calculations were below the recommended level. For P, there were no discernible differences between horses and ponies. There were also no clear effects of "inorganic" or "organic" P sources. The water solubility of "inorganic" sources was the key factor determining P metabolism. Water-soluble P sources exhibited higher true and apparent digestibility. The intake of these P sources led to hyperphosphatemia and hyperphosphaturia, even at low intakes. In other species, this has been shown to pose a health risk. Therefore, it is recommended to avoid the use of highly water-soluble "inorganic" P sources in horses and ponies. Given the lower digestibility of insoluble P sources, the factorially calculated P requirements for such sources are higher than the current recommendations.
Collapse
Affiliation(s)
- Isabelle Maier
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Schoenleutnerstr. 8, D-85764 Oberschleissheim, Germany;
| | | |
Collapse
|
2
|
Maekawa M, Maekawa T, Sasase T, Wakashima T, Uemura A, Uno K, Ohta T, Yamada T. Renal transcriptome analysis of uninephrectomized db/db mice identified a mechanism for the transition to severe diabetic nephropathy. Exp Anim 2024; 73:29-40. [PMID: 37482420 PMCID: PMC10877145 DOI: 10.1538/expanim.22-0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/09/2023] [Indexed: 07/25/2023] Open
Abstract
Diabetic nephropathy (DN), included in diabetic kidney disease (DKD), is a primary driver of end-stage renal disease (ESRD) leading to dialysis treatment. To develop new therapeutic drugs to prevent ESRD and avoid dialysis treatment, insight into DKD pathophysiology and animal models suitable for drug efficacy testing are needed. In this study, transcriptome analysis of kidneys from 26-week-old and 35-week-old uninephrectomized (UNX) db/db mice was used to identify the pathways that affect the deterioration of renal function in db/db mice. Differentially expressed genes suggested that there was increased interferon (IFN)-γ signaling during the 26 to 35-week period. Modules that changed between 26 and 35 weeks of age extracted by weighted gene co-expression network analysis (WGCNA) suggested increased the tumor necrosis factor (TNF)-α and nuclear factor-kappa B (NF-κB) signaling pathway in component cells of glomeruli. The protein-protein interaction (PPI) network analysis identified Cxcl16 as a hub gene for those signaling pathways, and it was shown that the pathways in this module changed when the glomerular filtration rate decreased in patients with DN. These results suggested the possibility that signaling mediated by Cxcl16 induced by IFN-γ and TNF-α between 26 and 35 weeks of age leads to renal fibrosis, resulting in severe disease. Drugs that target such pathways can be options for developing drugs for DN. We also think that the uninephrectomized db/db mouse can be used as an animal model of severe DKD and to evaluate efficacy in patients with DN.
Collapse
Affiliation(s)
- Mariko Maekawa
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan
| | - Tatsuya Maekawa
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Tomohiko Sasase
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Takeshi Wakashima
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Atsuhiro Uemura
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Kinuko Uno
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Takahisa Yamada
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan
| |
Collapse
|
3
|
Elsurer Afsar R, Afsar B, Ikizler TA. Fibroblast Growth Factor 23 and Muscle Wasting: A Metabolic Point of View. Kidney Int Rep 2023; 8:1301-1314. [PMID: 37441473 PMCID: PMC10334408 DOI: 10.1016/j.ekir.2023.04.027] [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: 12/19/2022] [Revised: 04/13/2023] [Accepted: 04/24/2023] [Indexed: 07/15/2023] Open
Abstract
Protein energy wasting (PEW), mostly characterized by decreased body stores of protein and energy sources, particularly in the skeletal muscle compartment, is highly prevalent in patients with moderate to advanced chronic kidney disease (CKD). Fibroblast growth factor 23 (FGF23) is an endocrine hormone secreted from bone and has systemic actions on skeletal muscle. In CKD, FGF23 is elevated and its coreceptor α-klotho is suppressed. Multiple lines of evidence suggest that FGF23 is interconnected with various mechanisms of skeletal muscle wasting in CKD, including systemic and local inflammation, exaggerated oxidative stress, insulin resistance (IR), and abnormalities in adipocytokine metabolism. Investigation of metabolic actions of FGF23 on muscle tissue could provide new insights into metabolic and nutritional abnormalities observed in patients with CKD.
Collapse
Affiliation(s)
- Rengin Elsurer Afsar
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Nephrology, Suleyman Demirel University Faculty of Medicine, Isparta, Turkey
| | - Baris Afsar
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Nephrology, Suleyman Demirel University Faculty of Medicine, Isparta, Turkey
| | - Talat Alp Ikizler
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt O’Brien Center for Kidney Disease, Nashville, Tennessee, USA
- Tennessee Valley Healthcare System, Nashville VA Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
4
|
Bollenbecker S, Czaya B, Gutiérrez OM, Krick S. Lung-kidney interactions and their role in chronic kidney disease-associated pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2022; 322:L625-L640. [PMID: 35272496 PMCID: PMC11684991 DOI: 10.1152/ajplung.00152.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic illnesses rarely present in a vacuum, devoid of other complications, and chronic kidney disease is hardly an exception. Comorbidities associated with chronic kidney disease lead to faster disease progression, expedited dialysis dependency, and a higher mortality rate. Although chronic kidney disease is most commonly accompanied by cardiovascular diseases and diabetes, there is clear cross talk between the lungs and kidneys pH balance, phosphate metabolism, and immune system regulation. Our present understanding of the exact underlying mechanisms that contribute to chronic kidney disease-related pulmonary disease is poor. This review summarizes the current research on kidney-pulmonary interorgan cross talk in the context of chronic kidney disease, highlighting various acute and chronic pulmonary diseases that lead to further complications in patient care. Treatment options for patients presenting with chronic kidney disease and lung disease are explored by assessing activated molecular pathways and the body's compensatory response mechanisms following homeostatic imbalance. Understanding the link between the lungs and kidneys will potentially improve health outcomes for patients and guide healthcare professionals to better understand how and when to treat each of the pulmonary comorbidities that can present with chronic kidney disease.
Collapse
Affiliation(s)
- Seth Bollenbecker
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Brian Czaya
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Orlando M Gutiérrez
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Stefanie Krick
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
5
|
Frangi G, Guicheteau M, Jacquot F, Pyka G, Kerckhofs G, Feyeux M, Veziers J, Guihard P, Halgand B, Sourice S, Guicheux J, Prieur X, Beck L, Beck-Cormier S. PiT2 deficiency prevents increase of bone marrow adipose tissue during skeletal maturation but not in OVX-induced osteoporosis. Front Endocrinol (Lausanne) 2022; 13:921073. [PMID: 36465661 PMCID: PMC9708882 DOI: 10.3389/fendo.2022.921073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
The common cellular origin between bone marrow adipocytes (BMAds) and osteoblasts contributes to the intimate link between bone marrow adipose tissue (BMAT) and skeletal health. An imbalance between the differentiation ability of BMSCs towards one of the two lineages occurs in conditions like aging or osteoporosis, where bone mass is decreased. Recently, we showed that the sodium-phosphate co-transporter PiT2/SLC20A2 is an important determinant for bone mineralization, strength and quality. Since bone mass is reduced in homozygous mutant mice, we investigated in this study whether the BMAT was also affected in PiT2-/- mice by assessing the effect of the absence of PiT2 on BMAT volume between 3 and 16 weeks, as well as in an ovariectomy-induced bone loss model. Here we show that the absence of PiT2 in juveniles leads to an increase in the BMAT that does not originate from an increased adipogenic differentiation of bone marrow stromal cells. We show that although PiT2-/- mice have higher BMAT volume than control PiT2+/+ mice at 3 weeks of age, BMAT volume do not increase from 3 to 16 weeks of age, leading to a lower BMAT volume in 16-week-old PiT2-/- compared to PiT2+/+ mice. In contrast, the absence of PiT2 does not prevent the increase in BMAT volume in a model of ovariectomy-induced bone loss. Our data identify SLC20a2/PiT2 as a novel gene essential for the maintenance of the BMAd pool in adult mice, involving mechanisms of action that remain to be elucidated, but which appear to be independent of the balance between osteoblastic and adipogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Giulia Frangi
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Marie Guicheteau
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Frederic Jacquot
- Nantes Université, CHU Nantes, Inserm, CNRS, CRCI2NA, Nantes, France
| | - Grzegorz Pyka
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UC Louvain, Louvain-la-Neuve, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UC Louvain, Louvain-la-Neuve, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
- IREC, Institute of Experimental and Clinical Research, UC Louvain, Woluwé-Saint-Lambert, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Magalie Feyeux
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Joëlle Veziers
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Pierre Guihard
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Boris Halgand
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Sophie Sourice
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Xavier Prieur
- Nantes Université, CNRS, Inserm, l’Institut du Thorax, Nantes, France
| | - Laurent Beck
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Sarah Beck-Cormier
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
- *Correspondence: Sarah Beck-Cormier,
| |
Collapse
|
6
|
Dobenecker B, Kienzle E, Siedler S. The Source Matters-Effects of High Phosphate Intake from Eight Different Sources in Dogs. Animals (Basel) 2021; 11:ani11123456. [PMID: 34944233 PMCID: PMC8698167 DOI: 10.3390/ani11123456] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/21/2023] Open
Abstract
Elevated serum phosphate concentrations are an established risk factor for cardiovascular disease and mortality in chronic kidney disease in various species. Independent associations of other parameters of phosphorus metabolism, such as phosphorus intake from different sources and serum concentrations of phosphorus, as well as parameters involved in the regulation, such as parathyroid hormone (PTH) or markers of bone turnover, have been studied in less detail. Therefore, the serum kinetics of phosphate, PTH, and the bone resorption marker bone-specific alkaline phosphatase (BAP) were investigated after 18 days of feeding a control diet and diets supplemented with eight different organic and inorganic phosphate sources aiming at 1.8% phosphorus per dry matter and calcium to phosphorus ratio between 1.3 and 1.7 to 1. Eight healthy beagle dogs (f/m, 2-4 years, 12.9 ± 1.4 kg body weight) were available for the trial. Highly significant differences in the serum kinetics of phosphorus, PTH, and BAP with the highest postprandial levels after feeding highly water-soluble sodium and potassium phosphates were found. We conclude that the use of certain inorganic phosphates in pet food is potentially harmful and should be restricted.
Collapse
|
7
|
Farooq A, Richman CM, Swain SM, Shahid RA, Vigna SR, Liddle RA. The Role of Phosphate in Alcohol-Induced Experimental Pancreatitis. Gastroenterology 2021; 161:982-995.e2. [PMID: 34051238 PMCID: PMC8380702 DOI: 10.1053/j.gastro.2021.05.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Heavy alcohol consumption is a common cause of acute pancreatitis; however, alcohol abuse does not always result in clinical pancreatitis. As a consequence, the factors responsible for alcohol-induced pancreatitis are not well understood. In experimental animals, it has been difficult to produce pancreatitis with alcohol. Clinically, alcohol use predisposes to hypophosphatemia, and hypophosphatemia has been observed in some patients with acute pancreatitis. Because of abundant protein synthesis, the pancreas has high metabolic demands, and reduced mitochondrial function leads to organelle dysfunction and pancreatitis. We proposed, therefore, that phosphate deficiency might limit adenosine triphosphate synthesis and thereby contribute to alcohol-induced pancreatitis. METHODS Mice were fed a low-phosphate diet (LPD) before orogastric administration of ethanol. Direct effects of phosphate and ethanol were evaluated in vitro in isolated mouse pancreatic acini. RESULTS LPD reduced serum phosphate levels. Intragastric administration of ethanol to animals maintained on an LPD caused severe pancreatitis that was ameliorated by phosphate repletion. In pancreatic acinar cells, low-phosphate conditions increased susceptibility to ethanol-induced cellular dysfunction through decreased bioenergetic stores, specifically affecting total cellular adenosine triphosphate and mitochondrial function. Phosphate supplementation prevented ethanol-associated cellular injury. CONCLUSIONS Phosphate status plays a critical role in predisposition to and protection from alcohol-induced acinar cell dysfunction and the development of acute alcohol-induced pancreatitis. This finding may explain why pancreatitis develops in only some individuals with heavy alcohol use and suggests a potential novel therapeutic approach to pancreatitis. Finally, an LPD plus ethanol provides a new model for studying alcohol-associated pancreatic injury.
Collapse
Affiliation(s)
- Ahmad Farooq
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Courtney M Richman
- School of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Sandip M Swain
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Rafiq A Shahid
- Department of Pathology, Brown University, Providence, Rhode Island
| | - Steven R Vigna
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Rodger A Liddle
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Veterans Affairs Health Care System, Durham, North Carolina.
| |
Collapse
|
8
|
Su X, Cheng Y, Zhang G, Wang B. Novel insights into the pathological mechanisms of metabolic related dyslipidemia. Mol Biol Rep 2021; 48:5675-5687. [PMID: 34218408 DOI: 10.1007/s11033-021-06529-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/27/2021] [Indexed: 12/21/2022]
Abstract
Due to the technological advances, it has been well-established that obesity is strongly correlated with various health problems. Among these problems, dyslipidemia is one of the most important concomitant symptoms under obese status which is the main driving force behind the pathological progression of cardio-metabolic disorder diseases. Importantly, the type of dyslipidemia, arising from concerted action of obesity, has been identified as "metabolic related dyslipidemia", which is characterized by increased circulating levels of Low density lipoprotein cholesterol (LDL-C), Triglycerides (TG) accompanied by lower circulating levels of High density lipoprotein cholesterol (HDL-C). On the other hand, the metabolic related dyslipidemia is being verified as a vital link between obesity and hypertension, diabetes mellitus, and Cardiovascular disease (CVD). In this review, we summarized the current understanding of metabolic related dyslipidemia and the potential mechanisms which lead to the pathogenesis of obesity. Meanwhile, we also summarized the emerging results which focused on several novel lipid bio-markers in metabolic related dyslipidemia, such as pro-protein convertase subtilisin/kexin type 9 (PCSK9) and sphingosine-1-phosphate (S1P), and their potential use as biomarkers of metabolic related dyslipidemia.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Ye Cheng
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China
| | - Guoming Zhang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| | - Bin Wang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, No. 2999 Jinshan Road, Xiamen, 361000, Fujian, China.
| |
Collapse
|
9
|
Su X, Chen X, Wang B. Pathology of metabolically-related dyslipidemia. Clin Chim Acta 2021; 521:107-115. [PMID: 34192528 DOI: 10.1016/j.cca.2021.06.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 12/29/2022]
Abstract
It is well established that overweight/obesity is closely associated with multiple health problems. Among these, dyslipidemia is the most important and main driving force behind pathologic development of cardio-metabolic disorders such as diabetes mellitus, atherosclerotic-related cardiovascular disease and hypertension. Notably, a subtype of dyslipidemia, metabolic related dyslipidemia, is now recognized as a vital link between obesity and multiple different cardiovascular diseases. This condition is characterized by increased low density lipoprotein cholesterol (LDL-C) and triglyceride (TG) and very low density lipoprotein cholesterol (VLDL-C) as well as decreased high density lipoprotein cholesterol (HDL-C) in serum. In this review, we summarize the current understanding of metabolic related dyslipidemia and the potential mechanisms which lead to the pathogenesis of obesity/overweight. We focus on several novel lipid biomarkers such as pro-protein convertase subtilisin/kexin type 9 (PCSK9) and sphingosine-1-phosphate (S1P) and their potential use as biomarkers of metabolic related dyslipidemia.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiang Chen
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Bin Wang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
10
|
Dobenecker B, Reese S, Herbst S. Effects of dietary phosphates from organic and inorganic sources on parameters of phosphorus homeostasis in healthy adult dogs. PLoS One 2021; 16:e0246950. [PMID: 33606750 PMCID: PMC7894875 DOI: 10.1371/journal.pone.0246950] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/28/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The impact of dietary phosphorus (P) excess, especially on renal and cardiovascular health, has been investigated in several species, but little is known in dogs. OBJECTIVE The aim of this study was to examine effects of different P sources on concentration and postprandial kinetics of selected parameters of P homeostasis in dogs. METHODS Eight beagles received one control diet (P 0.5% dry matter [DM]) and three high P diets (poultry meal, NaH2PO4, and KH2PO4; P 1.7% DM) for 18d. Urine samples were collected pre- and postprandially while faeces were collected quantitatively for 5d and analysed for minerals. On day 18, blood was sampled 1h pre- and 0.5, 1, 1.5, 2, 3, 5 and 7h postprandially. RESULTS Pi (KH2PO4, NaH2PO4) but not organic P caused an increased apparent P digestibility and significantly influenced kinetics of serum FGF23, parathyroid hormone, P, CrossLaps and bonespecific alkaline phosphatase, demonstrating a disrupted calcium (Ca) and P homeostasis with potential harm for renal, cardiovascular and skeletal health. CONCLUSIONS Results of feeding Pi to dogs indicate distinct disturbances of Ca and P metabolism, in contrast to organic sources. The use of Pi in food can therefore not be considered as safe. Further research, especially on dose and long-term effects, is warranted.
Collapse
Affiliation(s)
- Britta Dobenecker
- Chair of Animal Nutrition and Dietetics, Department of Animal Science, Ludwig-Maximilians- Universität, Munich, Germany
| | - Sven Reese
- Chair of Anatomy, Histology and Embryology, Department of Animal Science, Ludwig-Maximilians- Universität, Munich, Germany
| | - Sarah Herbst
- Chair of Animal Nutrition and Dietetics, Department of Animal Science, Ludwig-Maximilians- Universität, Munich, Germany
| |
Collapse
|
11
|
Su X, Peng D. Emerging functions of adipokines in linking the development of obesity and cardiovascular diseases. Mol Biol Rep 2020; 47:7991-8006. [PMID: 32888125 DOI: 10.1007/s11033-020-05732-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022]
Abstract
Increasing evidence shows that obesity is the critical factor in shaping cardio-metabolic phenotypes. However, the pathogenic mechanisms remain incompletely clarified. According to the published reports, adipose tissue communicates with several diverse organs, such as heart, lungs, and kidneys through the secretion of various cytokines named adipokines. The adipocytes isolated from obese mice or humans are dysfunctional with aberrant production of pro-inflammatory adipokines, which subsequently induce both acute and chronic inflammatory reaction and facilitate the process of cardio-metabolic disorder complications. Furthermore, the microenvironment within adipose tissue under obese status also influence the secretion of adipokines. Recently, given that several important adipokines have been completely researched and causally involved in various diseases, we could make a conclusion that adipokines play an essential role in modulating the development of cardio-metabolic disorder diseases, whereas several novel adipokines continue to be explored and elucidated. In the present review, we summarized the current knowledge of the microenvironment of adipose tissue and the published mechanisms whereby adipocytes affects obesity and cardiovascular diseases. On the other hand, we also provide the evidence to elucidate the functions of adipokines in controlling and regulating the inflammatory reactions which contribute to obesity and cardiovascular disease.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.,Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
12
|
Su X, Peng D. Adipokines as novel biomarkers of cardio-metabolic disorders. Clin Chim Acta 2020; 507:31-38. [PMID: 32283064 DOI: 10.1016/j.cca.2020.04.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/16/2022]
|
13
|
Bradham KD, Nelson CM, Diamond GL, Thayer WC, Scheckel KG, Noerpel M, Herbin-Davis K, Elek B, Thomas DJ. Dietary Lead and Phosphate Interactions Affect Oral Bioavailability of Soil Lead in the Mouse. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:12556-12564. [PMID: 31557437 PMCID: PMC8188726 DOI: 10.1021/acs.est.9b02803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Effects of dietary P level on the oral bioavailability of Pb present in soil were examined in a mouse model. Adult female C57BL/6 mice had free access to AIN-93G purified rodent diet amended with Pb as a soluble salt, Pb acetate, or in a soil matrix (NIST SRM 2710a). In these studies, the basal diet contained P at a nutritionally sufficient level (0.3% w/w) and the modified diets contained P at a lower (0.15%) or a higher (1.2%) level. For either dietary Pb source (Pb acetate or NIST SRM 2710a), low dietary P level markedly increased accumulation of Pb in bone, blood, and kidney. Tissue Pb levels in mice fed a high P in diet were not different from mice fed the basal P diet. Dietary P and Pb interacted to affect body weight change and feed efficiency in mice. The relative contribution of different Pb species in diet and feces was also affected by dietary P level. Differences in Pb species between diet and feces indicated that transformation of Pb species can occur during gastrointestinal tract transit. These interactions between Pb and P that alter Pb speciation may be important determinants of the bioavailability of Pb ingested in soil.
Collapse
Affiliation(s)
- Karen D Bradham
- Office of Research and Development , United States Environmental Protection Agency , Research Triangle Park , Durham , North Carolina 27711 , United States
| | - Clay M Nelson
- Office of Research and Development , United States Environmental Protection Agency , Research Triangle Park , Durham , North Carolina 27711 , United States
| | - Gary L Diamond
- SRC, Inc. , North Syracuse , New York 13212 , United States
| | | | - Kirk G Scheckel
- Office of Research and Development , United States Environmental Protection Agency , Cincinnati , Ohio 45224 , United States
| | - Matt Noerpel
- Office of Research and Development , United States Environmental Protection Agency , Cincinnati , Ohio 45224 , United States
| | - Karen Herbin-Davis
- Pharmacokinetics Branch, Integrated Systems Toxicology Division , National Health and Environmental Effects Laboratory, ORD, US EPA, RTP, NC , Durham , North Carolina 27709 , United States
| | - Brittany Elek
- Pharmacokinetics Branch, Integrated Systems Toxicology Division , National Health and Environmental Effects Laboratory, ORD, US EPA, RTP, NC , Durham , North Carolina 27709 , United States
| | - David J Thomas
- Pharmacokinetics Branch, Integrated Systems Toxicology Division , National Health and Environmental Effects Laboratory, ORD, US EPA, RTP, NC , Durham , North Carolina 27709 , United States
| |
Collapse
|
14
|
Hum JM, O'Bryan LM, Tatiparthi AK, Clinkenbeard EL, Ni P, Cramer MS, Bhaskaran M, Johnson RL, Wilson JM, Smith RC, White KE. Sustained Klotho delivery reduces serum phosphate in a model of diabetic nephropathy. J Appl Physiol (1985) 2019; 126:854-862. [PMID: 30605400 PMCID: PMC6485689 DOI: 10.1152/japplphysiol.00838.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/12/2018] [Accepted: 12/29/2018] [Indexed: 12/26/2022] Open
Abstract
Diabetic nephropathy (DN) is a primary cause of end-stage renal disease and is becoming more prevalent because of the global rise in type 2 diabetes. A model of DN, the db/db uninephrectomized ( db/db-uni) mouse, is characterized by obesity, as well as compromised renal function. This model also manifests defects in mineral metabolism common in DN, including hyperphosphatemia, which leads to severe endocrine disease. The FGF23 coreceptor, α-Klotho, circulates as a soluble, cleaved form (cKL) and may directly influence phosphate handling. Our study sought to test the effects of cKL on mineral metabolism in db/db-uni mice. Mice were placed into either mild or moderate disease groups on the basis of the albumin-to-creatinine ratio (ACR). Body weights of db/db-uni mice were significantly greater across the study compared with lean controls regardless of disease severity. Adeno-associated cKL administration was associated with increased serum Klotho, intact, bioactive FGF23 (iFGF23), and COOH-terminal fragments of FGF23 ( P < 0.05). Blood urea nitrogen was improved after cKL administration, and cKL corrected hyperphosphatemia in the high- and low-ACR db/db-uni groups. Interestingly, 2 wk after cKL delivery, blood glucose levels were significantly reduced in db/db-uni mice with high ACR ( P < 0.05). Interestingly, several genes associated with stabilizing active iFGF23 were also increased in the osteoblastic UMR-106 cell line with cKL treatment. In summary, delivery of cKL to a model of DN normalized blood phosphate levels regardless of disease severity, supporting the concept that targeting cKL-affected pathways could provide future therapeutic avenues in DN. NEW & NOTEWORTHY In this work, systemic and continuous delivery of the "soluble" or "cleaved" form of the FGF23 coreceptor α-Klotho (cKL) via adeno-associated virus to a rodent model of diabetic nephropathy (DN), the db/db uninephrectomized mouse, normalized blood phosphate levels regardless of disease severity. This work supports the concept that targeting cKL-affected pathways could provide future therapeutic avenues for the severe mineral metabolism defects associated with DN.
Collapse
Affiliation(s)
- Julia M Hum
- Division of Molecular Genetics and Gene Therapy, Department of Medical and Molecular Genetics, Indiana University School of Medicine , Indianapolis, Indiana
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University , Indianapolis, Indiana
| | - Linda M O'Bryan
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company , Indianapolis, Indiana
| | - Arun K Tatiparthi
- Lead Optimization Toxicology and Pharmacology, Covance Incorporated, Greenfield, Indiana
| | - Erica L Clinkenbeard
- Division of Molecular Genetics and Gene Therapy, Department of Medical and Molecular Genetics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Pu Ni
- Division of Molecular Genetics and Gene Therapy, Department of Medical and Molecular Genetics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Martin S Cramer
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company , Indianapolis, Indiana
| | - Manoj Bhaskaran
- Toxicology and Pathology, Eli Lilly and Company , Indianapolis, Indiana
| | - Robert L Johnson
- Toxicology and Pathology, Eli Lilly and Company , Indianapolis, Indiana
| | - Jonathan M Wilson
- Tailored Therapeutics, Eli Lilly and Company , Indianapolis, Indiana
| | - Rosamund C Smith
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company , Indianapolis, Indiana
| | - Kenneth E White
- Division of Molecular Genetics and Gene Therapy, Department of Medical and Molecular Genetics, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
15
|
Kétszeri M, Kirsch A, Frauscher B, Moschovaki-Filippidou F, Mooslechner AA, Kirsch AH, Schabhuettl C, Aringer I, Artinger K, Pregartner G, Ekart R, Breznik S, Hojs R, Goessler W, Schilcher I, Müller H, Obermayer-Pietsch B, Frank S, Rosenkranz AR, Eller P, Eller K. MicroRNA-142-3p improves vascular relaxation in uremia. Atherosclerosis 2019; 280:28-36. [PMID: 30453118 PMCID: PMC6591123 DOI: 10.1016/j.atherosclerosis.2018.11.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/31/2018] [Accepted: 11/09/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Chronic kidney disease (CKD) is strongly associated with a high burden of cardiovascular morbidity and mortality. Therefore, we aimed to characterize the putative role of microRNAs (miR)s in uremic vascular remodelling and endothelial dysfunction. METHODS We investigated the expression pattern of miRs in two independent end-stage renal disease (ESRD) cohorts and in the animal model of uremic DBA/2 mice via quantitative RT-PCR. Moreover, DBA/2 mice were treated with intravenous injections of synthetic miR-142-3p mimic and were analysed for functional and morphological vascular changes by mass spectrometry and wire myography. RESULTS The expression pattern of miRs was regulated in ESRD patients and was reversible after kidney transplantation. Out of tested miRs, only blood miR-142-3p was negatively associated with carotid-femoral pulse-wave velocity in CKD 5D patients. We validated these findings in a murine uremic model and found similar suppression of miR-142-3p as well as decreased acetylcholine-mediated vascular relaxation of the aorta. Therefore, we designed experiments to restore bioavailability of aortic miR-142-3p in vivo via intravenous injection of synthetic miR-142-3p mimic. This intervention restored acetylcholine-mediated vascular relaxation. CONCLUSIONS Taken together, we provide compelling evidence, both in humans and in mice, that miR-142-3p constitutes a potential pharmacological agent to prevent endothelial dysfunction and increased arterial stiffness in ESRD.
Collapse
Affiliation(s)
- Máté Kétszeri
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Andrijana Kirsch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria; Department of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Austria
| | - Bianca Frauscher
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | | | - Agnes A Mooslechner
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Alexander H Kirsch
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Corinna Schabhuettl
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Ida Aringer
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Katharina Artinger
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Clinical Centre Maribor, Slovenia; Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Silva Breznik
- Department of Radiology, University Clinical Centre Maribor, Slovenia
| | - Radovan Hojs
- Faculty of Medicine, University of Maribor, Maribor, Slovenia; Department of Nephrology, Clinic for Internal Medicine, University Clinical Center Maribor, Slovenia
| | - Walter Goessler
- Institute of Chemistry Analytical Chemistry, Karl-Franzens University Graz, Graz, Austria
| | - Irene Schilcher
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | - Helmut Müller
- Department of Surgery, Division of Transplantation Surgery, Medical University of Graz, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | - Alexander R Rosenkranz
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Austria.
| | - Kathrin Eller
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| |
Collapse
|
16
|
Imi Y, Yabiki N, Abuduli M, Masuda M, Yamanaka-Okumura H, Taketani Y. High phosphate diet suppresses lipogenesis in white adipose tissue. J Clin Biochem Nutr 2018. [PMID: 30487667 DOI: 10.3164/jcbn.17.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Excessive phosphate intake has been positively associated with renal and vascular dysfunction, conversely negatively associated with body fat accumulation. We investigated the effect of a high-phosphate diet on the expression of lipid metabolic genes in white adipose tissue and liver. Male 8-week-old Sprague-Dawley rats were fed a control diet containing 0.6% phosphate or a high-phosphate diet containing 1.5% phosphate for 4 weeks. In comparison to the control group, the HP group showed a significantly lower body fat mass and fasting plasma insulin level alongside decreased lipogenic and increased lipolytic gene expression in visceral fat. Additionally, the expression of genes involved in hepatic lipogenesis, hepatic glycogenesis, and triglyceride accumulation decreased in the high-phosphate group. Exogenous phosphate, parathyroid hormone, and fibroblast growth factor 23 did not directly affect the expression of lipolytic or lipogenic genes in 3T3-L1 adipocytes and HepG2 hepatocytes. Thus, the high-phosphate diet suppressed the activity of white adipose tissue by increasing lipolytic gene expression and decreasing lipogenic gene expression. These effects could have been caused by the lowered fasting plasma insulin level that occurred in response to the high-phosphate diet, but were not directly caused by the increases in plasma phosphate or phosphaturic hormones.
Collapse
Affiliation(s)
- Yukiko Imi
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Norie Yabiki
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maerjianghan Abuduli
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
17
|
Frauscher B, Kirsch AH, Schabhüttl C, Schweighofer K, Kétszeri M, Pollheimer M, Dragun D, Schröder K, Rosenkranz AR, Eller K, Eller P. Autophagy Protects From Uremic Vascular Media Calcification. Front Immunol 2018; 9:1866. [PMID: 30154792 PMCID: PMC6102358 DOI: 10.3389/fimmu.2018.01866] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease and diabetes mellitus are associated with extensive media calcification, which leads to increased cardiovascular morbidity and mortality. Here, we investigated the role of autophagy in the pathogenesis of uremic vascular media calcification. DBA/2 mice were fed with high-phosphate diet (HPD) in order to cause vascular calcification. DBA/2 mice on standard chow diet were used as control. In parallel, autophagy and its response to rapamycin, 3-methyladenine (3-MA), and bafilomycin were studied in an in vitro model using mouse vascular smooth muscle cells (MOVAS). DBA/2 mice on HPD developed severe vascular media calcification, which is mirrored in vitro by culturing MOVAS under calcifying conditions. Both, in vitro and in vivo, autophagy significantly increased in MOVAS under calcifying conditions and in aortas of HPD mice, respectively. Histologically, autophagy was located to the aortic Tunica media, but also vascular endothelial cells, and was found to continuously increase during HPD treatment. 3-MA as well as bafilomycin blocked autophagy in MOVAS and increased calcification. Vice versa, rapamycin treatment further increased autophagy and resulted in a significant decrease of vascular calcification in vitro and in vivo. Rapamycin reduced Runx2 transcription levels in aortas and MOVAS to control levels, whereas it increased α-smooth muscle actin and Sm22α transcription in MOVAS to control levels. Furthermore, rapamycin-treated HPD mice survived significantly longer compared to HPD controls. These findings indicate that autophagy is an endogenous response of vascular smooth muscle cells (VSMC) to protect from calcification in uremia. Induction of autophagy by rapamycin protects cells and mice from uremic media calcification possibly by inhibiting osteogenic transdifferentiation of VSMC.
Collapse
Affiliation(s)
- Bianca Frauscher
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Alexander H Kirsch
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Corinna Schabhüttl
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Kerstin Schweighofer
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Máté Kétszeri
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Duska Dragun
- Clinic for Nephrology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Freie Universitat Berlin, Humboldt-Universitat zu Berlin, Berlin, Germany.,Institute of Health (BIH), Berlin, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Alexander R Rosenkranz
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
18
|
Imi Y, Yabiki N, Abuduli M, Masuda M, Yamanaka-Okumura H, Taketani Y. High phosphate diet suppresses lipogenesis in white adipose tissue. J Clin Biochem Nutr 2018; 63:181-191. [PMID: 30487667 PMCID: PMC6252294 DOI: 10.3164/jcbn.17-141] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 01/31/2018] [Indexed: 02/06/2023] Open
Abstract
Excessive phosphate intake has been positively associated with renal and vascular dysfunction, conversely negatively associated with body fat accumulation. We investigated the effect of a high-phosphate diet on the expression of lipid metabolic genes in white adipose tissue and liver. Male 8-week-old Sprague-Dawley rats were fed a control diet containing 0.6% phosphate or a high-phosphate diet containing 1.5% phosphate for 4 weeks. In comparison to the control group, the HP group showed a significantly lower body fat mass and fasting plasma insulin level alongside decreased lipogenic and increased lipolytic gene expression in visceral fat. Additionally, the expression of genes involved in hepatic lipogenesis, hepatic glycogenesis, and triglyceride accumulation decreased in the high-phosphate group. Exogenous phosphate, parathyroid hormone, and fibroblast growth factor 23 did not directly affect the expression of lipolytic or lipogenic genes in 3T3-L1 adipocytes and HepG2 hepatocytes. Thus, the high-phosphate diet suppressed the activity of white adipose tissue by increasing lipolytic gene expression and decreasing lipogenic gene expression. These effects could have been caused by the lowered fasting plasma insulin level that occurred in response to the high-phosphate diet, but were not directly caused by the increases in plasma phosphate or phosphaturic hormones.
Collapse
Affiliation(s)
- Yukiko Imi
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Norie Yabiki
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maerjianghan Abuduli
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
19
|
Frauscher B, Artinger K, Kirsch AH, Aringer I, Moschovaki-Filippidou F, Kétszeri M, Schabhüttl C, Rainer PP, Schmidt A, Stojakovic T, Fahrleitner-Pammer A, Rosenkranz AR, Eller P, Eller K. A New Murine Model of Chronic Kidney Disease-Mineral and Bone Disorder. Int J Endocrinol 2017; 2017:1659071. [PMID: 29387084 PMCID: PMC5745741 DOI: 10.1155/2017/1659071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/31/2017] [Accepted: 09/11/2017] [Indexed: 01/18/2023] Open
Abstract
Chronic kidney disease (CKD) is associated with mineral and bone disorder (MBD), which is the main cause of the extensively increased cardiovascular mortality in the CKD population. We now aimed to establish a new murine experimental CKD-MBD model. Dilute brown non-Agouti (DBA/2) mice were fed with high-phosphate diet for 4 (HPD4) or 7 (HPD7) days, then with standard chow diet (SCD) and subsequently followed until day 84. They were compared to DBA/2 mice maintained on SCD during the whole study period. Both 4 and 7 days HPD-fed mice developed phosphate nephropathy with tubular atrophy, interstitial fibrosis, decreased glomerular filtration rate, and increased serum urea levels. The abdominal aorta of HPD-treated mice showed signs of media calcification. Histomorphometric analysis of HPD-treated mice showed decreased bone volume/tissue volume, low mineral apposition rate, and low bone formation rate as compared to SCD-fed mice, despite increased parathyroid hormone levels. Overall, the observed phenotype was more pronounced in the HPD7 group. In summary, we established a new, noninvasive, and therefore easy to perform reproducible CKD-MBD model, which showed media calcification, secondary hyperparathyroidism, and low-turnover bone disease.
Collapse
Affiliation(s)
- Bianca Frauscher
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Katharina Artinger
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | | | - Ida Aringer
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | | | - Máté Kétszeri
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Corinna Schabhüttl
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Peter P. Rainer
- Clinical Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Albrecht Schmidt
- Clinical Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | | | | | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| |
Collapse
|
20
|
Phosphorus Supplementation Recovers the Blunted Diet-Induced Thermogenesis of Overweight and Obese Adults: A Pilot Study. Nutrients 2016; 8:nu8120801. [PMID: 27941661 PMCID: PMC5188456 DOI: 10.3390/nu8120801] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/27/2016] [Accepted: 12/06/2016] [Indexed: 11/17/2022] Open
Abstract
Diet-induced thermogenesis (DIT) is believed to be largely related to ATP production, which is dependent on phosphorus (P) availability. We aimed to test the effect of P addition on DIT of lean and overweight/obese healthy subjects. DIT was measured with or without P in 10 lean and 13 overweight/obese adults in a double-blind randomized cross-over pilot study with one week washout period. After 10 h overnight fast, resting metabolic rate, respiratory quotient, and substrate utilization were measured at fasting and every 30 min for 3 h after subjects drank a standardized glucose solution, with P (500 mg) or placebo pills. Subjective ratings of hunger and satiety were assessed before and after the end of each experiment using validated visual analogue scale (VAS) questionnaires. Overweight/obese subjects had a blunted DIT with placebo, while P supplementation induced a 23% increase in their DIT area under the curve (p < 0.05), which was associated with a significant increase in carbohydrate oxidation. Subjects had lower appetite following P supplementation, which was expressed as a significantly (p = 0.02) lower desire to eat a meal (4.0 ± 0.7 cm) compared with placebo (5.8 ± 0.9 cm). P supplementation recovers the blunted diet-induced thermogenesis in overweight and obese subjects and enhances their postprandial satiety.
Collapse
|
21
|
Kirsch AH, Kirsch A, Artinger K, Schabhüttl C, Goessler W, Klymiuk I, Gülly C, Fritz GA, Frank S, Wimmer R, Brodmann M, Anders HJ, Pramstaller PP, Rosenkranz AR, Eller K, Eller P. Heterogeneous susceptibility for uraemic media calcification and concomitant inflammation within the arterial tree. Nephrol Dial Transplant 2015; 30:1995-2005. [PMID: 26185049 PMCID: PMC4656037 DOI: 10.1093/ndt/gfv265] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/01/2015] [Indexed: 12/27/2022] Open
Abstract
Background End-stage renal disease (ESRD) is strongly associated with arterial calcification of the tunica media, decreased vascular compliance and sudden cardiac death. Here, we analysed the distribution pattern of uraemic media calcification and concomitant inflammation in mice and men. Methods Uraemia was induced in DBA/2 mice with high-phosphate diet. Subsequently, we analysed arterial medial calcification using histology, mass spectrometry, and wire myography. Gene expression was quantified using a whole transcriptome array and quantitative PCR. In a cohort of 36 consecutive patients with CKD stage 4–5, we measured the calcium score of the coronary arteries, the ascending thoracic aorta and the infrarenal abdominal aorta using computed tomography scans. Results Uraemic DBA/2 mice showed only minor calcifications in thoracic aortas, whereas there was overt media calcification in abdominal aortas. The transcriptional profile and immunohistochemistry revealed induction of Vcam1 expression by vascular smooth muscle cells in uraemic abdominal aortas. Macrophages infiltrated the tunica media of the abdominal aorta. Anti-inflammatory treatment did not improve uraemic media calcification in our animal model. Arterial calcifications in ESRD patients showed a similar distribution pattern in computed tomography scans, with higher calcium scores of the abdominal aorta when compared with the thoracic aorta. Conclusion Taken together, there was a similar heterogeneous pattern of calcification in both mice and humans, where the abdominal aorta was more prone to media calcification when compared with the thoracic aorta. In uraemia, smooth muscle cells of the abdominal aorta showed a phenotypic switch to an inflammatory and osteoblastic phenotype.
Collapse
Affiliation(s)
- Alexander H Kirsch
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Andrijana Kirsch
- Center of Molecular Medicine, Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Katharina Artinger
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Corinna Schabhüttl
- Department of Internal Medicine, Division of Angiology, Medical University of Graz, Graz, Austria
| | - Walter Goessler
- Institute of Chemistry-Analytical Chemistry, Karl-Franzens University of Graz, Graz, Austria
| | - Ingeborg Klymiuk
- Center for Medical Research, Core Facility Molecular Biology, Medical University of Graz, Graz, Austria
| | - Christian Gülly
- Center for Medical Research, Core Facility Molecular Biology, Medical University of Graz, Graz, Austria
| | - Gerald A Fritz
- Department of Radiology, Medical University of Graz, Graz, Austria
| | - Saša Frank
- Center of Molecular Medicine, Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Roxana Wimmer
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Marianne Brodmann
- Department of Internal Medicine, Division of Angiology, Medical University of Graz, Graz, Austria
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München-Innenstadt, Munich, Germany
| | | | - Alexander R Rosenkranz
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- Department of Internal Medicine, Division of Angiology, Medical University of Graz, Graz, Austria Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Graz, Austria
| |
Collapse
|
22
|
Hoffmanová I, Kraml P, Anděl M. Renal risk associated with sodium phosphate medication: safe in healthy individuals, potentially dangerous in others. Expert Opin Drug Saf 2015; 14:1097-110. [DOI: 10.1517/14740338.2015.1044970] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Liang L, Zheng C, Zhang H, Xu S, Zhang Z, Hu C, Bi L, Fan Z, Han B, Xu W. Exploring type II microcalcifications in benign and premalignant breast lesions by shell-isolated nanoparticle-enhanced Raman spectroscopy (SHINERS). SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2014; 132:397-402. [PMID: 24887501 DOI: 10.1016/j.saa.2014.04.147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 04/17/2014] [Accepted: 04/19/2014] [Indexed: 05/14/2023]
Abstract
The characteristics of type II microcalcifications in fibroadenoma (FB), atypical ductal hyperplasia (ADH), and ductal carcinoma in situ (DCIS) breast tissues has been analyzed by the fingerprint features of Raman spectroscopy. Fresh breast tissues were first handled to frozen sections and then they were measured by normal Raman spectroscopy. Due to inherently low sensitivity of Raman scattering, Au@SiO2 shell-isolated nanoparticle-enhanced Raman spectroscopy (SHINERS) technique was utilized. A total number of 71 Raman spectra and 70 SHINERS spectra were obtained from the microcalcifications in benign and premalignant breast tissues. Principal component analysis (PCA) was used to distinguish the type II microcalcifications between these tissues. This is the first time to detect type II microcalcifications in premalignant (ADH and DCIS) breast tissue frozen sections, and also the first time SHINERS has been utilized for breast cancer detection. Conclusions demonstrated in this paper confirm that SHINERS has great potentials to be applied to the identification of breast lesions as an auxiliary method to mammography in the early diagnosis of breast cancer.
Collapse
Affiliation(s)
- Lijia Liang
- State Key Laboratory for Supramolecular Structure and Materials, Jilin University, Changchun 130012, China
| | - Chao Zheng
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Haipeng Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Shuping Xu
- State Key Laboratory for Supramolecular Structure and Materials, Jilin University, Changchun 130012, China
| | - Zhe Zhang
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Chengxu Hu
- State Key Laboratory for Supramolecular Structure and Materials, Jilin University, Changchun 130012, China
| | - Lirong Bi
- Pathology Department, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Bing Han
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun 130021, China.
| | - Weiqing Xu
- State Key Laboratory for Supramolecular Structure and Materials, Jilin University, Changchun 130012, China.
| |
Collapse
|
24
|
Kirsch AH, Smaczny N, Riegelbauer V, Sedej S, Hofmeister A, Stojakovic T, Goessler W, Brodmann M, Pilger E, Rosenkranz AR, Eller K, Eller P. Regulatory T cells improve nephrocalcinosis but not dystrophic cardiac calcinosis in DBA/2 mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:382-90. [PMID: 23746654 DOI: 10.1016/j.ajpath.2013.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/17/2013] [Accepted: 04/24/2013] [Indexed: 12/24/2022]
Abstract
Nephrocalcinosis is characterized by aberrant deposition of calcium in the kidneys and is seen in phosphate nephropathy, primary hyperparathyroidism, and distal renal tubular acidosis. To further evaluate the specific pathophysiologic role of T cells in ectopic calcification, we used DBA/2 mice that are prone to develop nephrocalcinosis and dystrophic cardiac calcinosis. Female DBA/2 mice were depleted of T cells (n = 10) or regulatory T cells (Tregs) (n = 15) using either an anti-CD3ɛ or an anti-CD25 monoclonal antibody and compared with isotype-treated controls (n = 9; n = 15), respectively. After this immunomodulation, the DBA/2 mice were given a high-phosphate diet for 9 days and the degree of calcification was assessed by microcomputed tomography. Successful depletion was confirmed by flow cytometry of splenocytes. In DBA/2 mice, the high-phosphate diet induced a phenotype of nephrocalcinosis and dystrophic cardiac calcinosis. T-cell depletion significantly increased renal calcification in microcomputed tomography (P = 0.022). Concordantly, Treg depletion significantly deteriorated acute phosphate nephropathy (P = 0.039) and was associated with a significantly increased mortality rate (P = 0.004). Immunomodulation had no impact on the amount of cardiac calcification. Semiquantitative histopathologic evaluations with Alizarin Red staining independently confirmed the respective radiologic measurements. In summary, our data suggest a pivotal role of T cells, particularly Tregs, in the progression of nephrocalcinosis and emphasize the fact that inflammation deteriorates the outcome in acute phosphate nephropathy.
Collapse
Affiliation(s)
- Alexander H Kirsch
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ribitsch W, Rosenkranz AR. Prehypertension and prechronic kidney disease: fact or fiction? Expert Rev Cardiovasc Ther 2011; 9:651-3. [DOI: 10.1586/erc.11.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|