1
|
Feenstra L, Reijrink M, Pasch A, Smith ER, Visser LM, Bulthuis M, Lodewijk ME, Mastik MF, Greuter MJW, Slart RHJA, Mulder DJ, Pol RA, te Velde-Keyzer CA, Krenning G, Hillebrands JL. Calciprotein particle counts associate with vascular remodelling in chronic kidney disease. Cardiovasc Res 2024; 120:1953-1966. [PMID: 39102822 PMCID: PMC11629976 DOI: 10.1093/cvr/cvae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/09/2024] [Accepted: 06/01/2024] [Indexed: 08/07/2024] Open
Abstract
AIMS Calciprotein particles (CPPs) are circulating calcium and phosphate nanoparticles associated with the development of vascular calcification (VC) in chronic kidney disease (CKD). Although recent studies have been focusing on associations of CPPs with the presence of VC in CKD, insights in the underlying processes and mechanisms by which CPPs might aggravate VC and vascular dysfunction in vivo are currently lacking. Here, we assessed the overall burden of abdominal VC in healthy kidney donors and CKD patients and subsequently performed transcriptome profiling in the vascular tissue obtained from these subjects, linking outcome to CPP counts and calcification propensity. METHODS AND RESULTS Calcification scores were quantified in renal arteries, iliac arteries, and abdominal aorta using computed tomography (CT) scans of kidney donors and CKD patients. The vascular tissue was collected from kidney donors (renal artery) and CKD patients (iliac artery), after which bulk RNA sequencing and gene set enrichment analysis (GSEA) were performed on a subset of patients. Calcification propensity (crystallization time, T50) was measured using nephelometry and CPP counts with microparticle flow cytometric analysis. Increased calcification scores (based on CT) were found in CKD patients compared to kidney donors. Transcriptome profiling revealed enrichment for processes related to endothelial activation, inflammation, extracellular matrix (ECM) remodelling, and ossification in CKD vascular biopsies compared to kidney donors. Calcification propensity was increased in CKD, as well as CPP counts, with the latter being significantly associated with markers of vascular remodelling. CONCLUSION Our findings reveal that CKD is characterized by systemic VC with increased calcification propensity and CPP counts. Transcriptome profiling showed altered vascular gene expression with enrichment for endothelial activation, inflammation, ECM remodelling, and ossification. Moreover, we demonstrate, for the first time, that vascular remodelling processes are associated with increased circulating CPP counts. Interventions targeting CPPs are promising avenues for alleviating vascular remodelling and VC in CKD.
Collapse
Affiliation(s)
- Lian Feenstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Melanie Reijrink
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Internal Medicine, Division of Vascular Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Edward R Smith
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Lotte M Visser
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Marian Bulthuis
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Monique E Lodewijk
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Mirjam F Mastik
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Marcel J W Greuter
- Department of Radiology, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Riemer H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Douwe J Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert A Pol
- Department of Vascular and Transplant Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Charlotte A te Velde-Keyzer
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Guido Krenning
- Department of Clinical Pharmacy and Pharmacology, Division Experimental Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
2
|
Elliott J, Tang PK. Fibroblast Growth Factor 23 - a review with particular reference to the physiology and pathophysiology of phosphate homeostasis in the cat. Vet J 2024:106271. [PMID: 39608700 DOI: 10.1016/j.tvjl.2024.106271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/08/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Fibroblast growth factor-23 (FGF23) is a phosphaturic hormone, discovery of which has transformed our understanding of mineral regulation in healthy mammals, including the cat. It is produced by osteoblasts and osteocytes and its prime role is to regulate phosphate entry into extracellular fluid (from bone and via the gut) and its excretion via the kidney. It interacts with other hormones (calcitriol and parathyroid hormone), inhibiting their activation and secretion respectively and so impacts on calcium as well as phosphate homeostasis. Physiological factors regulating its secretion are not well understood, although phosphate ion sensing is likely to be important. Calcium and magnesium ions are also involved and unravelling the control points and integration of the system regulating bone turnover and mineral balance whilst preventing soft tissue (non-osseous) mineralisation is a future research goal. Calciprotein particle size and number likely play an important role in this system but precisely how remains to be determined. Elevated serum FGF23 is the earliest indicator of mineral bone disorder associated with chronic kidney disease in human patients and in cats, enabling reference-range serum phosphorus to be maintained despite reduction in glomerular filtration rate which limits phosphate excretion. FGF23 also predicts CKD progression and survival in cats. The many factors influencing its secretion at different stages of CKD, including relative iron deficiency, anaemia and chronic systemic inflammation, hypomagnesaemia and α-klotho deficiency are discussed in this review, where the data available in cats with naturally occurring CKD is presented alongside that from rodent models and human CKD patients.
Collapse
Affiliation(s)
- Jonathan Elliott
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU.
| | - Pak Kan Tang
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU
| |
Collapse
|
3
|
Stepanov A, Shishkova D, Markova V, Markova Y, Frolov A, Lazebnaya A, Oshchepkova K, Perepletchikova D, Smirnova D, Basovich L, Repkin E, Kutikhin A. Proteomic Profiling of Endothelial Cell Secretomes After Exposure to Calciprotein Particles Reveals Downregulation of Basement Membrane Assembly and Increased Release of Soluble CD59. Int J Mol Sci 2024; 25:11382. [PMID: 39518935 PMCID: PMC11546392 DOI: 10.3390/ijms252111382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Calciprotein particles (CPPs) are essential circulating scavengers of excessive Ca2+ and PO43- ions, representing a vehicle that removes them from the human body and precludes extraskeletal calcification. Having been internalised by endothelial cells (ECs), CPPs induce their dysfunction, which is accompanied by a remarkable molecular reconfiguration, although little is known about this process's extracellular signatures. Here, we applied ultra-high performance liquid chromatography-tandem mass spectrometry to perform a secretome-wide profiling of the cell culture supernatant from primary human coronary artery ECs (HCAECs) and internal thoracic artery ECs (HITAECs) treated with primary CPPs (CPP-P), secondary CPPs (CPP-S), magnesiprotein particles (MPPs), or Ca2+/Mg2+-free Dulbecco's phosphate-buffered saline (DPBS) for 24 h. Incubation with CPP-P/CPP-S significantly altered the profiles of secreted proteins, delineating physiological and pathological endothelial secretomes. Neither pathway enrichment analysis nor the interrogation of protein-protein interactions detected extracellular matrix- and basement membrane-related molecular terms in the protein datasets from CPP-P/CPP-S-treated ECs. Both proteomic profiling and enzyme-linked immunosorbent assay identified an increased level of protectin (CD59) and reduced levels of osteonectin (SPARC), perlecan (HSPG2), and fibronectin (FN1) in the cell culture supernatant upon CPP-P/CPP-S treatment. Elevated soluble CD59 and decreased release of basement membrane components might be considered as potential signs of dysfunctional endothelium.
Collapse
Affiliation(s)
- Alexander Stepanov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Yulia Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Alexey Frolov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Anastasia Lazebnaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Karina Oshchepkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| | - Daria Perepletchikova
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia; (D.P.); (D.S.); (L.B.)
| | - Daria Smirnova
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia; (D.P.); (D.S.); (L.B.)
| | - Liubov Basovich
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia; (D.P.); (D.S.); (L.B.)
| | - Egor Repkin
- Resource Centre for Molecular and Cell Technologies, St. Petersburg State University, Universitetskaya Embankment, 7/9, 199034 St. Petersburg, Russia;
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, 650002 Kemerovo, Russia; (A.S.); (D.S.); (V.M.); (Y.M.); (A.F.); (A.L.); (K.O.)
| |
Collapse
|
4
|
Chiang HJ, Peng HH, Weng KF, Hsiung KC, Liang CY, Kuo SL, Ojcius DM, Young JDE, Shih SR. Mineralo-organic particles inhibit influenza A virus infection by targeting viral hemagglutinin activity. Nanomedicine (Lond) 2024; 19:2375-2390. [PMID: 39320315 PMCID: PMC11492690 DOI: 10.1080/17435889.2024.2403326] [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: 05/25/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Aim: Mineralo-organic particles, naturally present in human body fluids, participate in ectopic calcification and inflammatory diseases. These particles coexist with influenza A virus (IAV) in the same microenvironment during viral infection. Our objective was to investigate the functional consequences of the potential interactions between these particles and the virions.Materials & methods: We used in vitro models, including electron microscopy, fluorescence microscopy, hemagglutination assay and viral infection assays to examine the interactions.Results: Mineralo-organic particles bind to IAV virions through interactions involving particle-bound fetuin-A and mineral content, effectively engaging viral hemagglutinin. These interactions result in hindered viral infection.Conclusion: These findings uncover the novel interactions between mineralo-organic particles and IAV, highlighting the impact of virus microenvironment complexity.
Collapse
Affiliation(s)
- Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Hsin-Hsin Peng
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan, 33302, Taiwan
- Division of Chinese Medicine Obstetrics & Gynecology, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
| | - Kuo-Feng Weng
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Microbiology & Immunology, Stanford University SOM, Stanford, CA94305, USA
| | - Kuei-Ching Hsiung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Chieh-Yu Liang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO63110, USA
| | - Shun-Li Kuo
- Division of Chinese Medicine Obstetrics & Gynecology, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - David M. Ojcius
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan, 33302, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA94103, USA
| | | | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food & Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science & Technology, Taoyuan, 33303, Taiwan
| |
Collapse
|
5
|
Lafage-Proust MH, Magne D. Biology of bone mineralization and ectopic calcifications: the same actors for different plays. Arch Pediatr 2024; 31:4S3-4S12. [PMID: 39343471 DOI: 10.1016/s0929-693x(24)00151-9] [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] [Indexed: 10/01/2024]
Abstract
Bone has several crucial functions. It is essential for locomotion and allows our body to stand erect against gravity. A mismatch between the mechanical stresses applied to it and its mechanical resistance leads to fractures. Bone also has numerous endocrine functions. It acts as a reservoir for minerals such as calcium and phosphorus, making it the target of calciotropic hormones that mobilize these minerals, particularly calcium, according to the body's needs. Additionally, bone secretes hormones, notably fibroblast growth factor 23 (FGF23), which regulates urinary excretion of phosphate and the bioavailability of active vitamin D. Bone mineralization is the process that facilitates the organized deposition of minerals in the bone matrix, providing rigidity and appropriate mechanical resistance. This process is compromised in genetically related bone mineralization disorders, such as those causing hypophosphatemia or hypophosphatasia. Conversely, calcification can be pathological, affecting soft tissues like the blood vessels, as seen in generalized arterial calcification of infancy (GACI) or arterial calcification due to CD73 deficiency (ACDC). The aim of this article is to first present the composition and structure of the mineralized bone matrix, to review the current understanding of the molecular mechanisms of mineralization, and finally to discuss the conditions associated with ectopic calcification and the underlying mechanisms.
Collapse
Affiliation(s)
| | - David Magne
- University of Lyon I; ICBMS, UMR CNRS 5246, F-69622, LYON, France.
| |
Collapse
|
6
|
Mencke R, Al Ali L, de Koning MSLY, Pasch A, Minnion M, Feelisch M, van Veldhuisen DJ, van der Horst ICC, Gansevoort RT, Bakker SJL, de Borst MH, van Goor H, van der Harst P, Lipsic E, Hillebrands JL. Serum Calcification Propensity Is Increased in Myocardial Infarction and Hints at a Pathophysiological Role Independent of Classical Cardiovascular Risk Factors. Arterioscler Thromb Vasc Biol 2024; 44:1884-1894. [PMID: 38899469 DOI: 10.1161/atvbaha.124.320974] [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: 03/20/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Vascular calcification is associated with increased mortality in patients with cardiovascular disease. Secondary calciprotein particles are believed to play a causal role in the pathophysiology of vascular calcification. The maturation time (T50) of calciprotein particles provides a measure of serum calcification propensity. We compared T50 between patients with ST-segment-elevated myocardial infarction and control subjects and studied the association of T50 with cardiovascular risk factors and outcome. METHODS T50 was measured by nephelometry in 347 patients from the GIPS-III trial (Metabolic Modulation With Metformin to Reduce Heart Failure After Acute Myocardial Infarction: Glycometabolic Intervention as Adjunct to Primary Coronary Intervention in ST Elevation Myocardial Infarction: a Randomized Controlled Trial) and in 254 matched general population controls from PREVEND (Prevention of Renal and Vascular End-Stage Disease). We also assessed the association between T50 and left ventricular ejection fraction, as well as infarct size, the incidence of ischemia-driven reintervention during 5 years of follow-up, and serum nitrite as a marker of endothelial dysfunction. RESULTS Patients with ST-segment-elevated myocardial infarction had a significantly lower T50 (ie, higher serum calcification propensity) compared with controls (T50: 289±63 versus 338±56 minutes; P<0.001). In patients with ST-segment-elevated myocardial infarction, lower T50 was associated with female sex, lower systolic blood pressure, lower total cholesterol, lower LDL (low-density lipoprotein) cholesterol, lower triglycerides, and higher HDL (high-density lipoprotein) cholesterol but not with circulating nitrite or nitrate. Ischemia-driven reintervention was associated with higher LDL (P=0.03) and had a significant interaction term for T50 and sex (P=0.005), indicating a correlation between ischemia-driven reintervention and T50 above the median in men and below the median in women, between 150 days and 5 years of follow-up. CONCLUSIONS Serum calcification propensity is increased in patients with ST-segment-elevated myocardial infarction compared with the general population, and its contribution is more pronounced in women than in men. Its lack of/inverse association with nitrite and blood pressure confirms T50 to be orthogonal to traditional cardiovascular disease risk factors. Lower T50 was associated with a more favorable serum lipid profile, suggesting the involvement of divergent pathways of calcification stress and lipid stress in the pathophysiology of myocardial infarction.
Collapse
Affiliation(s)
- Rik Mencke
- Department of Pathology and Medical Biology, Division of Pathology (R.M., H.v.G., J.L.H.), University Medical Center Groningen, the Netherlands
| | - Lawien Al Ali
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | - Marie-Sophie L Y de Koning
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland (A.P.)
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Austria (A.P.)
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and University Hospital Southampton NHS Foundation Trust, United Kingdom (M.M., M.F.)
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and University Hospital Southampton NHS Foundation Trust, United Kingdom (M.M., M.F.)
| | - Dirk J van Veldhuisen
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | | | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology (R.T.G., S.J.L.B., M.H.d.B.), University Medical Center Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology (R.T.G., S.J.L.B., M.H.d.B.), University Medical Center Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology (R.T.G., S.J.L.B., M.H.d.B.), University Medical Center Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology (R.M., H.v.G., J.L.H.), University Medical Center Groningen, the Netherlands
| | - Pim van der Harst
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, the Netherlands (P.v.d.H.)
| | - Erik Lipsic
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology (R.M., H.v.G., J.L.H.), University Medical Center Groningen, the Netherlands
| |
Collapse
|
7
|
Klein GL. Phosphate as an adjunct to calcium in promoting coronary vascular calcification in chronic inflammatory states. eLife 2024; 13:e91808. [PMID: 38864841 PMCID: PMC11168742 DOI: 10.7554/elife.91808] [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: 08/11/2023] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
Bone releases calcium and phosphate in response to pro-inflammatory cytokine-mediated inflammation. The body develops impaired urinary excretion of phosphate with age and chronic inflammation given the reduction of the kidney protein Klotho, which is essential to phosphate excretion. Phosphate may also play a role in the development of the resistance of the parathyroid calcium-sensing receptor (CaSR) to circulating calcium thus contributing to calcium retention in the circulation. Phosphate can contribute to vascular smooth muscle dedifferentiation with manifestation of osteoblastogenesis and ultimately endovascular calcium phosphate precipitation. Thus phosphate, along with calcium, contributes to the calcification and inflammation of atherosclerotic plaques and the origin of these elements is likely the bone, which serves as storage for the majority of the body's supply of extracellular calcium and phosphate. Early cardiac evaluation of patients with chronic inflammation and attempts at up-regulating the parathyroid CaSR with calcimimetics or introducing earlier anti-resorptive treatment with bone active pharmacologic agents may serve to delay onset or reduce the quantity of atherosclerotic plaque calcification in these patients.
Collapse
Affiliation(s)
- Gordon L Klein
- Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical BranchGalvestonUnited States
| |
Collapse
|
8
|
Zeper LW, Bos C, Leermakers PA, Franssen GM, Raavé R, Hoenderop JGJ, de Baaij JHF. Liver and spleen predominantly mediate calciprotein particle clearance in a rat model of chronic kidney disease. Am J Physiol Renal Physiol 2024; 326:F622-F634. [PMID: 38420675 DOI: 10.1152/ajprenal.00239.2023] [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: 08/11/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Calciprotein particles (CPPs) provide an efficient mineral buffering system to prevent the complexation of phosphate and calcium in the circulation. However, in chronic kidney disease (CKD), the phosphate load exceeds the mineral buffering capacity, resulting in the formation of crystalline CPP2 particles. CPP2 have been associated with cardiovascular events and mortality. Moreover, CPP2 have been demonstrated to induce calcification in vitro. In this study, we examined the fate of CPP2 in a rat model of CKD. Calcification was induced in Sprague-Dawley rats by 5/6 nephrectomy (5/6-Nx) combined with a high-phosphate diet. Control rats received sham surgery and high-phosphate diet. Twelve weeks after surgery, kidney failure was significantly induced in 5/6-Nx rats as determined by enhanced creatinine and urea plasma levels and abnormal kidney histological architecture. Subsequently, radioactive and fluorescent (FITC)-labeled CPP2 ([89Zr]Zr-CPP2-FITC) were injected intravenously to determine clearance in vivo. Using positron emission tomography scans and radioactive biodistribution measurements, it was demonstrated that [89Zr]Zr-CPP2-FITC are mainly present in the liver and spleen in both 5/6-Nx and sham rats. Immunohistochemistry showed that [89Zr]Zr-CPP2-FITC are predominantly taken up by Kupffer cells and macrophages. However, [89Zr]Zr-CPP2-FITC could also be detected in hepatocytes. In the different parts of the aorta and in the blood, low values of [89Zr]Zr-CPP2-FITC were detectable, independent of the presence of calcification. CPP2 are cleared rapidly from the circulation by the liver and spleen in a rat model of CKD. In the liver, Kupffer cells, macrophages, and hepatocytes contribute to CPP2 clearance.NEW & NOTEWORTHY Calciprotein particles (CPPs) buffer calcium and phosphate in the blood to prevent formation of crystals. In CKD, increased phosphate levels may exceed the buffering capacity of CPPs, resulting in crystalline CPPs that induce calcification. This study demonstrates that labeled CPPs are predominantly cleared from the circulation in the liver by Kupffer cells, macrophages, and hepatocytes. Our results suggest that targeting liver CPP clearance may reduce the burden of crystalline CPP in the development of vascular calcification.
Collapse
Affiliation(s)
- Lara W Zeper
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caro Bos
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter A Leermakers
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben M Franssen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René Raavé
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Shishkova D, Lobov A, Repkin E, Markova V, Markova Y, Sinitskaya A, Sinitsky M, Kondratiev E, Torgunakova E, Kutikhin A. Calciprotein Particles Induce Cellular Compartment-Specific Proteome Alterations in Human Arterial Endothelial Cells. J Cardiovasc Dev Dis 2023; 11:5. [PMID: 38248875 PMCID: PMC10816121 DOI: 10.3390/jcdd11010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Calciprotein particles (CPPs) are indispensable scavengers of excessive Ca2+ and PO43- ions in blood, being internalised and recycled by liver and spleen macrophages, monocytes, and endothelial cells (ECs). Here, we performed a pathway enrichment analysis of cellular compartment-specific proteomes in primary human coronary artery ECs (HCAEC) and human internal thoracic artery ECs (HITAEC) treated with primary (amorphous) or secondary (crystalline) CPPs (CPP-P and CPPs, respectively). Exposure to CPP-P and CPP-S induced notable upregulation of: (1) cytokine- and chemokine-mediated signaling, Ca2+-dependent events, and apoptosis in cytosolic and nuclear proteomes; (2) H+ and Ca2+ transmembrane transport, generation of reactive oxygen species, mitochondrial outer membrane permeabilisation, and intrinsic apoptosis in the mitochondrial proteome; (3) oxidative, calcium, and endoplasmic reticulum (ER) stress, unfolded protein binding, and apoptosis in the ER proteome. In contrast, transcription, post-transcriptional regulation, translation, cell cycle, and cell-cell adhesion pathways were underrepresented in cytosol and nuclear compartments, whilst biosynthesis of amino acids, mitochondrial translation, fatty acid oxidation, pyruvate dehydrogenase activity, and energy generation were downregulated in the mitochondrial proteome of CPP-treated ECs. Differentially expressed organelle-specific pathways were coherent in HCAEC and HITAEC and between ECs treated with CPP-P or CPP-S. Proteomic analysis of mitochondrial and nuclear lysates from CPP-treated ECs confirmed bioinformatic filtration findings.
Collapse
Affiliation(s)
- Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Arseniy Lobov
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia;
| | - Egor Repkin
- Centre for Molecular and Cell Technologies, St. Petersburg State University, Universitetskaya Embankment, 7/9, 199034 St. Petersburg, Russia;
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Yulia Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Anna Sinitskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Maxim Sinitsky
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Egor Kondratiev
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Evgenia Torgunakova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| |
Collapse
|
10
|
Kuro-O M. Calcium phosphate microcrystallopathy as a paradigm of chronic kidney disease progression. Curr Opin Nephrol Hypertens 2023; 32:344-351. [PMID: 37074676 PMCID: PMC10242516 DOI: 10.1097/mnh.0000000000000890] [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] [Indexed: 04/20/2023]
Abstract
PURPOSE OF REVIEW Calciprotein particles (CPP) are colloidal mineral-protein complexes mainly composed of solid-phase calcium phosphate and serum protein fetuin-A. CPP appear in the blood and renal tubular fluid after phosphate intake, playing critical roles in (patho)physiology of mineral metabolism and chronic kidney disease (CKD). This review aims at providing an update of current knowledge on CPP. RECENT FINDINGS CPP formation is regarded as a defense mechanism against unwanted growth of calcium phosphate crystals in the blood and urine. CPP are polydisperse colloids and classified based on the density and crystallinity of calcium phosphate. Low-density CPP containing amorphous (noncrystalline) calcium phosphate function as an inducer of FGF23 expression in osteoblasts and a carrier of calcium phosphate to the bone. However, once transformed to high-density CPP containing crystalline calcium phosphate, CPP become cytotoxic and inflammogenic, inducing cell death in renal tubular cells, calcification in vascular smooth muscle cells, and innate immune responses in macrophages. SUMMARY CPP potentially behave like a pathogen that causes renal tubular damage, chronic inflammation, and vascular calcification. CPP have emerged as a promising therapeutic target for CKD and cardiovascular complications.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Antiaging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
11
|
Zeper LW, Smith ER, Ter Braake AD, Tinnemans PT, de Baaij JHF, Hoenderop JGJ. Calciprotein Particle Synthesis Strategy Determines In Vitro Calcification Potential. Calcif Tissue Int 2023; 112:103-117. [PMID: 36326853 PMCID: PMC9813048 DOI: 10.1007/s00223-022-01036-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
Circulating calciprotein particles (CPP), colloids of calcium, phosphate and proteins, were identified as potential drivers of the calcification process in chronic kidney disease. The present study compared CPP produced using different protocols with respect to particle morphology, composition, particle number and in vitro calcification potency. CPP were synthesized with 4.4 mM (CPP-A and B) or 6 mM (CPP-C and D) phosphate and 2.8 mM (CPP-A and B) or 10 mM (CPP-C and D) calcium, with either bovine fetuin-A (CPP-C) or fetal bovine serum (CPP-A, B and D) as a source of protein, and incubated for 7 (CPP-A2) or 14 days (CPP-B2), 12 h (CPP-C2, D2 and B1) or 30 min (CPP-D1). Particle number was determined with nanoparticle tracking and calcium content was measured in CPP preparations and to determine human vascular smooth muscle cell (hVSMC) calcification. Morphologically, CPP-C2 were the largest. Particle number did not correspond to the calcium content of CPP. Both methods of quantification resulted in variable potencies of CPP2 to calcify VSMC, with CPP-B2 as most stable inducer of hVSMC calcification. In contrast, CPP-B1 and D1 were unable to induce calcification of hVSMC, and endogenous CPP derived from pooled serum of dialysis patients were only able to calcify hVSMC to a small extent compared to CPP2.CPP synthesized using different protocols appear morphologically similar, but in vitro calcification potency is dependent on composition and how the CPP are quantified. Synthetic CPP are not comparable to endogenous CPP in terms of the calcification propensity.
Collapse
Affiliation(s)
- Lara W Zeper
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Nephrology, University of Melbourne, Parkville, VIC, Australia
| | - Anique D Ter Braake
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Paul T Tinnemans
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Sheng W, Ji G, Zhang L. Role of macrophage scavenger receptor MSR1 in the progression of non-alcoholic steatohepatitis. Front Immunol 2022; 13:1050984. [PMID: 36591228 PMCID: PMC9797536 DOI: 10.3389/fimmu.2022.1050984] [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: 09/22/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the progressive form of nonalcoholic fatty liver disease (NAFLD), and the dysregulation of lipid metabolism and oxidative stress are the typical features. Subsequent dyslipidemia and oxygen radical production may render the formation of modified lipids. Macrophage scavenger receptor 1 (MSR1) is responsible for the uptake of modified lipoprotein and is one of the key molecules in atherosclerosis. However, the unrestricted uptake of modified lipoproteins by MSR1 and the formation of cholesterol-rich foamy macrophages also can be observed in NASH patients and mouse models. In this review, we highlight the dysregulation of lipid metabolism and oxidative stress in NASH, the alteration of MSR1 expression in physiological and pathological conditions, the formation of modified lipoproteins, and the role of MSR1 on macrophage foaming and NASH development and progression.
Collapse
|
13
|
Shishkova D, Lobov A, Zainullina B, Matveeva V, Markova V, Sinitskaya A, Velikanova E, Sinitsky M, Kanonykina A, Dyleva Y, Kutikhin A. Calciprotein Particles Cause Physiologically Significant Pro-Inflammatory Response in Endothelial Cells and Systemic Circulation. Int J Mol Sci 2022; 23:ijms232314941. [PMID: 36499266 PMCID: PMC9738209 DOI: 10.3390/ijms232314941] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Calciprotein particles (CPPs) represent an inherent mineral buffering system responsible for the scavenging of excessive Ca2+ and PO43- ions in order to prevent extraskeletal calcification, although contributing to the development of endothelial dysfunction during the circulation in the bloodstream. Here, we performed label-free proteomic profiling to identify the functional consequences of CPP internalisation by endothelial cells (ECs) and found molecular signatures of significant disturbances in mitochondrial and lysosomal physiology, including oxidative stress, vacuolar acidification, accelerated proteolysis, Ca2+ cytosolic elevation, and mitochondrial outer membrane permeabilisation. Incubation of intact ECs with conditioned medium from CPP-treated ECs caused their pro-inflammatory activation manifested by vascular cell adhesion molecule 1 (VCAM1) and intercellular adhesion molecule 1 (ICAM1) upregulation and elevated release of interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1/ C-C motif ligand 2 (MCP-1/CCL2). Among the blood cells, monocytes were exclusively responsible for CPP internalisation. As compared to the co-incubation of donor blood with CPPs in the flow culture system, intravenous administration of CPPs to Wistar rats caused a considerably higher production of chemokines, indicating the major role of monocytes in CPP-triggered inflammation. Upregulation of sICAM-1 and IL-8 also suggested a notable contribution of endothelial dysfunction to systemic inflammatory response after CPP injections. Collectively, our results demonstrate the pathophysiological significance of CPPs and highlight the need for the development of anti-CPP therapies.
Collapse
Affiliation(s)
- Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Arseniy Lobov
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia
| | - Bozhana Zainullina
- Centre for Molecular and Cell Technologies, St. Petersburg State University, Universitetskaya Embankment, 7/9, 199034 St. Petersburg, Russia
| | - Vera Matveeva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anna Sinitskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Elena Velikanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Maxim Sinitsky
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anastasia Kanonykina
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Yulia Dyleva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
- Correspondence: ; Tel.: +7-960-907-7067
| |
Collapse
|
14
|
Gudgeon J, Marín-Rubio JL, Trost M. The role of macrophage scavenger receptor 1 (MSR1) in inflammatory disorders and cancer. Front Immunol 2022; 13:1012002. [PMID: 36325338 PMCID: PMC9618966 DOI: 10.3389/fimmu.2022.1012002] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/28/2022] [Indexed: 08/27/2023] Open
Abstract
Macrophage scavenger receptor 1 (MSR1), also named CD204, holds key inflammatory roles in multiple pathophysiologic processes. Present primarily on the surface of various types of macrophage, this receptor variably affects processes such as atherosclerosis, innate and adaptive immunity, lung and liver disease, and more recently, cancer. As highlighted throughout this review, the role of MSR1 is often dichotomous, being either host protective or detrimental to the pathogenesis of disease. We will discuss the role of MSR1 in health and disease with a focus on the molecular mechanisms influencing MSR1 expression, how altered expression affects disease process and macrophage function, the limited cell signalling pathways discovered thus far, the emerging role of MSR1 in tumour associated macrophages as well as the therapeutic potential of targeting MSR1.
Collapse
Affiliation(s)
| | - José Luis Marín-Rubio
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Matthias Trost
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
15
|
Bogdanova M, Zabirnyk A, Malashicheva A, Semenova D, Kvitting JPE, Kaljusto ML, Perez MDM, Kostareva A, Stensløkken KO, Sullivan GJ, Rutkovskiy A, Vaage J. Models and Techniques to Study Aortic Valve Calcification in Vitro, ex Vivo and in Vivo. An Overview. Front Pharmacol 2022; 13:835825. [PMID: 35721220 PMCID: PMC9203042 DOI: 10.3389/fphar.2022.835825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aortic valve stenosis secondary to aortic valve calcification is the most common valve disease in the Western world. Calcification is a result of pathological proliferation and osteogenic differentiation of resident valve interstitial cells. To develop non-surgical treatments, the molecular and cellular mechanisms of pathological calcification must be revealed. In the current overview, we present methods for evaluation of calcification in different ex vivo, in vitro and in vivo situations including imaging in patients. The latter include echocardiography, scanning with computed tomography and magnetic resonance imaging. Particular emphasis is on translational studies of calcific aortic valve stenosis with a special focus on cell culture using human primary cell cultures. Such models are widely used and suitable for screening of drugs against calcification. Animal models are presented, but there is no animal model that faithfully mimics human calcific aortic valve disease. A model of experimentally induced calcification in whole porcine aortic valve leaflets ex vivo is also included. Finally, miscellaneous methods and aspects of aortic valve calcification, such as, for instance, biomarkers are presented.
Collapse
Affiliation(s)
- Maria Bogdanova
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arsenii Zabirnyk
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Daria Semenova
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Mari-Liis Kaljusto
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Woman and Children Health, Karolinska Institute, Stockholm, Sweden
| | - Kåre-Olav Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
16
|
Li M, Wang ZW, Fang LJ, Cheng SQ, Wang X, Liu NF. Programmed cell death in atherosclerosis and vascular calcification. Cell Death Dis 2022; 13:467. [PMID: 35585052 PMCID: PMC9117271 DOI: 10.1038/s41419-022-04923-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022]
Abstract
The concept of cell death has been expanded beyond apoptosis and necrosis to additional forms, including necroptosis, pyroptosis, autophagy, and ferroptosis. These cell death modalities play a critical role in all aspects of life, which are noteworthy for their diverse roles in diseases. Atherosclerosis (AS) and vascular calcification (VC) are major causes for the high morbidity and mortality of cardiovascular disease. Despite considerable advances in understanding the signaling pathways associated with AS and VC, the exact molecular basis remains obscure. In the article, we review the molecular mechanisms that mediate cell death and its implications for AS and VC. A better understanding of the mechanisms underlying cell death in AS and VC may drive the development of promising therapeutic strategies.
Collapse
Affiliation(s)
- Min Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Zhen-Wei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Li-Juan Fang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Shou-Quan Cheng
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Xin Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
17
|
Tiong MK, Cai MMX, Toussaint ND, Tan SJ, Pasch A, Smith ER. Effect of nutritional calcium and phosphate loading on calciprotein particle kinetics in adults with normal and impaired kidney function. Sci Rep 2022; 12:7358. [PMID: 35513558 PMCID: PMC9072391 DOI: 10.1038/s41598-022-11065-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/31/2022] [Indexed: 12/20/2022] Open
Abstract
Plasma approaches metastability with respect to its calcium and phosphate content, with only minor perturbations in ionic activity needed to sustain crystal growth once nucleated. Physiologically, calcium and phosphate are intermittently absorbed from the diet each day, yet plasma concentrations of these ions deviate minimally post-prandially. This implies the existence of a blood-borne mineral buffer system to sequester calcium phosphates and minimise the risk of deposition in the soft tissues. Calciprotein particles (CPP), endogenous mineral-protein colloids containing the plasma protein fetuin-A, may fulfill this function but definitive evidence linking dietary mineral loading with their formation is lacking. Here we demonstrate that CPP are formed as a normal physiological response to feeding in healthy adults and that this occurs despite minimal change in conventional serum mineral markers. Further, in individuals with Chronic Kidney Disease (CKD), in whom mineral handling is impaired, we show that both fasting and post-prandial levels of CPP precursors are markedly augmented and strongly inversely correlated with kidney function. This study highlights the important, but often neglected, contribution of colloidal biochemistry to mineral homeostasis and provides novel insight into the dysregulation of mineral metabolism in CKD.
Collapse
Affiliation(s)
- Mark K Tiong
- Department of Nephrology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC, 3052, Australia. .,Department of Medicine (RMH), University of Melbourne, Parkville, Australia.
| | - Michael M X Cai
- Department of Nephrology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC, 3052, Australia
| | - Nigel D Toussaint
- Department of Nephrology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC, 3052, Australia.,Department of Medicine (RMH), University of Melbourne, Parkville, Australia
| | - Sven-Jean Tan
- Department of Nephrology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC, 3052, Australia.,Department of Medicine (RMH), University of Melbourne, Parkville, Australia
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland.,Lindenhofspital Bern, Bern, Switzerland.,Department of Physiology and Pathophysiology, Johannes Kepler University, Linz, Austria
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC, 3052, Australia. .,Department of Medicine (RMH), University of Melbourne, Parkville, Australia.
| |
Collapse
|
18
|
Rudloff S, Jahnen-Dechent W, Huynh-Do U. Tissue chaperoning—the expanded functions of fetuin-A beyond inhibition of systemic calcification. Pflugers Arch 2022; 474:949-962. [PMID: 35403906 PMCID: PMC8995415 DOI: 10.1007/s00424-022-02688-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023]
Abstract
AbstractTraditionally, fetuin-A embodies the prototype anti-calcification protein in the blood, preventing cardiovascular calcification. Low serum fetuin-A is generally associated with mineralization dysbalance and enhanced mortality in end stage renal disease. Recent evidence indicates that fetuin-A is a crucial factor moderating tissue inflammation and fibrosis, as well as a systemic indicator of acute inflammatory disease. Here, the expanded function of fetuin-A is discussed in the context of mineralization and inflammation biology. Unbalanced depletion of fetuin-A in this context may be the critical event, triggering a vicious cycle of progressive calcification, inflammation, and tissue injury. Hence, we designate fetuin-A as tissue chaperone and propose the potential use of exogenous fetuin-A as prophylactic agent or emergency treatment in conditions that are associated with acute depletion of endogenous protein.
Collapse
Affiliation(s)
- Stefan Rudloff
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen, University Medical Faculty, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland.
| |
Collapse
|
19
|
Birukov A, Polemiti E, Jäger S, Stefan N, Schulze MB. Fetuin-A and risk of diabetes-related vascular complications: a prospective study. Cardiovasc Diabetol 2022; 21:6. [PMID: 34998417 PMCID: PMC8742328 DOI: 10.1186/s12933-021-01439-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Fetuin-A is a hepatokine which has the capacity to prevent vascular calcification. Moreover, it is linked to the induction of metabolic dysfunction, insulin resistance and associated with increased risk of diabetes. It has not been clarified whether fetuin-A associates with risk of vascular, specifically microvascular, complications in patients with diabetes. We aimed to investigate whether pre-diagnostic plasma fetuin-A is associated with risk of complications once diabetes develops. METHODS Participants with incident type 2 diabetes and free of micro- and macrovascular disease from the European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam cohort (n = 587) were followed for microvascular and macrovascular complications (n = 203 and n = 60, respectively, median follow-up: 13 years). Plasma fetuin-A was measured approximately 4 years prior to diabetes diagnosis. Prospective associations between baseline fetuin-A and risk of complications were assessed with Cox regression. RESULTS In multivariable models, fetuin-A was linearly inversely associated with incident total and microvascular complications, hazard ratio (HR, 95% CI) per standard deviation (SD) increase: 0.86 (0.74; 0.99) for total, 0.84 (0.71; 0.98) for microvascular and 0.92 (0.68; 1.24) for macrovascular complications. After additional adjustment for cardiometabolic plasma biomarkers, including triglycerides and high-density lipoprotein, the associations were slightly attenuated: 0.88 (0.75; 1.02) for total, 0.85 (0.72; 1.01) for microvascular and 0.95 (0.67; 1.34) for macrovascular complications. No interaction by sex could be observed (p > 0.10 for all endpoints). CONCLUSIONS Our data show that lower plasma fetuin-A levels measured prior to the diagnosis of diabetes may be etiologically implicated in the development of diabetes-associated microvascular disease.
Collapse
Affiliation(s)
- Anna Birukov
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany.
| | - Elli Polemiti
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Norbert Stefan
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, Tübingen, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany.
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
20
|
Leifheit-Nestler M, Vogt I, Haffner D, Richter B. Phosphate Is a Cardiovascular Toxin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:107-134. [DOI: 10.1007/978-3-030-91623-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Shishkova DK, Velikanova EA, Bogdanov LA, Sinitsky MY, Kostyunin AE, Tsepokina AV, Gruzdeva OV, Mironov AV, Mukhamadiyarov RA, Glushkova TV, Krivkina EO, Matveeva VG, Hryachkova ON, Markova VE, Dyleva YA, Belik EV, Frolov AV, Shabaev AR, Efimova OS, Popova AN, Malysheva VY, Kolmykov RP, Sevostyanov OG, Russakov DM, Dolganyuk VF, Gutakovsky AK, Zhivodkov YA, Kozhukhov AS, Brusina EB, Ismagilov ZR, Barbarash OL, Yuzhalin AE, Kutikhin AG. Calciprotein Particles Link Disturbed Mineral Homeostasis with Cardiovascular Disease by Causing Endothelial Dysfunction and Vascular Inflammation. Int J Mol Sci 2021; 22:ijms222212458. [PMID: 34830334 PMCID: PMC8626027 DOI: 10.3390/ijms222212458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
An association between high serum calcium/phosphate and cardiovascular events or death is well-established. However, a mechanistic explanation of this correlation is lacking. Here, we examined the role of calciprotein particles (CPPs), nanoscale bodies forming in the human blood upon its supersaturation with calcium and phosphate, in cardiovascular disease. The serum of patients with coronary artery disease or cerebrovascular disease displayed an increased propensity to form CPPs in combination with elevated ionised calcium as well as reduced albumin levels, altogether indicative of reduced Ca2+-binding capacity. Intravenous administration of CPPs to normolipidemic and normotensive Wistar rats provoked intimal hyperplasia and adventitial/perivascular inflammation in both balloon-injured and intact aortas in the absence of other cardiovascular risk factors. Upon the addition to primary human arterial endothelial cells, CPPs induced lysosome-dependent cell death, promoted the release of pro-inflammatory cytokines, stimulated leukocyte adhesion, and triggered endothelial-to-mesenchymal transition. We concluded that CPPs, which are formed in the blood as a result of altered mineral homeostasis, cause endothelial dysfunction and vascular inflammation, thereby contributing to the development of cardiovascular disease.
Collapse
Affiliation(s)
- Daria K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Elena A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Leo A. Bogdanov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Maxim Yu. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Alexander E. Kostyunin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Anna V. Tsepokina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Olga V. Gruzdeva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Andrey V. Mironov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Rinat A. Mukhamadiyarov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Tatiana V. Glushkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Evgenia O. Krivkina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Vera G. Matveeva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Oksana N. Hryachkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Victoria E. Markova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Yulia A. Dyleva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Ekaterina V. Belik
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Alexey V. Frolov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Amin R. Shabaev
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Olga S. Efimova
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Anna N. Popova
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Valentina Yu. Malysheva
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Roman P. Kolmykov
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Oleg G. Sevostyanov
- Institute of Fundamental Sciences, Kemerovo State University, 6 Krasnaya Street, 650000 Kemerovo, Russia; (O.G.S.); (D.M.R.); (V.F.D.)
| | - Dmitriy M. Russakov
- Institute of Fundamental Sciences, Kemerovo State University, 6 Krasnaya Street, 650000 Kemerovo, Russia; (O.G.S.); (D.M.R.); (V.F.D.)
| | - Viatcheslav F. Dolganyuk
- Institute of Fundamental Sciences, Kemerovo State University, 6 Krasnaya Street, 650000 Kemerovo, Russia; (O.G.S.); (D.M.R.); (V.F.D.)
| | - Anton K. Gutakovsky
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of the Russian Academy of Sciences, 13 Akademika Lavrentieva Avenue, 630090 Novosibirsk, Russia; (A.K.G.); (Y.A.Z.); (A.S.K.)
| | - Yuriy A. Zhivodkov
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of the Russian Academy of Sciences, 13 Akademika Lavrentieva Avenue, 630090 Novosibirsk, Russia; (A.K.G.); (Y.A.Z.); (A.S.K.)
| | - Anton S. Kozhukhov
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of the Russian Academy of Sciences, 13 Akademika Lavrentieva Avenue, 630090 Novosibirsk, Russia; (A.K.G.); (Y.A.Z.); (A.S.K.)
| | - Elena B. Brusina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Zinfer R. Ismagilov
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Olga L. Barbarash
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Arseniy E. Yuzhalin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Anton G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
- Correspondence: ; Tel.: +7-960-907-7067
| |
Collapse
|
22
|
Abstract
Chronic kidney disease (CKD) has reached epidemic proportions globally. The natural course of chronic kidney disease is almost uniformly progressive, albeit at different rates in different individuals. The downhill course appears to pervade kidney diseases of all etiologies and seems to spiral down a self-perpetuating vortex, even if the original insult is ameliorated or controlled. In this issue of the JCI, Shiizaki, Tsubouchi, and colleagues proposed a model of renal tubule luminal calcium phosphate crystallopathy that accounts for renal function demise. Calcium phosphate crystals attached to TLR4 and underwent endocytosis at the brush border, triggering inflammation and fibrosis. This mechanism might operate in different kinds of kidney disease, with a theoretical phosphate concentration threshold in the proximal tubular lumen, beyond which is triggered undesirable downstream effects that eventuate in loss of renal function. If this model parallels human CKD, clinicians may focus efforts on determining phosphate exposure in the proximal tubular lumen.
Collapse
|
23
|
Kutikhin AG, Feenstra L, Kostyunin AE, Yuzhalin AE, Hillebrands JL, Krenning G. Calciprotein Particles: Balancing Mineral Homeostasis and Vascular Pathology. Arterioscler Thromb Vasc Biol 2021; 41:1607-1624. [PMID: 33691479 PMCID: PMC8057528 DOI: 10.1161/atvbaha.120.315697] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anton G. Kutikhin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Lian Feenstra
- Department of Pathology and Medical Biology, Division of Pathology (L.F., J.-L.H.), University Medical Center Groningen, University of Groningen, the Netherlands
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology (L.F., G.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Alexander E. Kostyunin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Arseniy E. Yuzhalin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology (L.F., J.-L.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology (L.F., G.K.), University Medical Center Groningen, University of Groningen, the Netherlands
- Sulfateq B.V., Admiraal de Ruyterlaan 5, 9726 GN, Groningen, the Netherlands (G.K.)
| |
Collapse
|
24
|
Koeppert S, Ghallab A, Peglow S, Winkler CF, Graeber S, Büscher A, Hengstler JG, Jahnen-Dechent W. Live Imaging of Calciprotein Particle Clearance and Receptor Mediated Uptake: Role of Calciprotein Monomers. Front Cell Dev Biol 2021; 9:633925. [PMID: 33996793 PMCID: PMC8116800 DOI: 10.3389/fcell.2021.633925] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Background The liver-derived plasma protein fetuin A is a systemic inhibitor of ectopic calcification. Fetuin-A stabilizes calcium phosphate mineral initially as ion clusters to form calciprotein monomers (CPM), and then as larger multimeric consolidations containing amorphous calcium phosphate (primary CPP, CPP 1) or more crystalline phases (secondary CPP, CPP 2). CPM and CPP mediate excess mineral stabilization, transport and clearance from circulation. Methods We injected i.v. synthetic fluorescent CPM and studied their clearance by live two-photon microscopy. We analyzed organ sections by fluorescence microscopy to assess CPM distribution. We studied cellular clearance and cytotoxicity by flow cytometry and live/dead staining, respectively, in cultured macrophages, liver sinusoidal endothelial cells (LSEC), and human proximal tubule epithelial HK-2 cells. Inflammasome activation was scored in macrophages. Fetuin A monomer and CPM charge were analyzed by ion exchange chromatography. Results Live mice cleared CPP in the liver as published previously. In contrast, CPM were filtered by kidney glomeruli into the Bowman space and the proximal tubules, suggesting tubular excretion of CPM-bound calcium phosphate and reabsorption of fetuin A. Fetuin-A monomer clearance was negligible in liver and low in kidney. Anion exchange chromatography revealed that fetuin A monomer was negatively charged, whereas CPM appeared neutral, suggesting electrochemical selectivity of CPM versus fetuin A. CPM were non-toxic in any of the investigated cell types, whereas CPP 1 were cytotoxic. Unlike CPP, CPM also did not activate the inflammasome. Conclusions Fetuin-A prevents calcium phosphate precipitation by forming CPM, which transform into CPP. Unlike CPP, CPM do not trigger inflammation. CPM are readily cleared in the kidneys, suggesting CPM as a physiological transporter of excess calcium and phosphate. Upon prolonged circulation, e.g., in chronic kidney disease, CPM will coalesce and form CPP, which cannot be cleared by the kidney, but will be endocytosed by liver sinusoidal endothelial cells and macrophages. Large amounts of CPP trigger inflammation. Chronic CPM and CPP clearance deficiency thus cause calcification by CPP deposition in blood vessels and soft tissues, as well as inflammation.
Collapse
Affiliation(s)
- Sina Koeppert
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany.,Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Sarah Peglow
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | | | - Steffen Graeber
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Andrea Büscher
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Jan Georg Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
25
|
Uedono H, Mori K, Ochi A, Nakatani S, Miki Y, Tsuda A, Morioka T, Nagata Y, Imanishi Y, Shoji T, Inaba M, Emoto M. Effects of fetuin-A-containing calciprotein particles on posttranslational modifications of fetuin-A in HepG2 cells. Sci Rep 2021; 11:7486. [PMID: 33820929 PMCID: PMC8021573 DOI: 10.1038/s41598-021-86881-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 03/15/2021] [Indexed: 01/06/2023] Open
Abstract
Fetuin-A is an inhibitor of ectopic calcification that is expressed mainly in hepatocytes and is secreted into the circulation after posttranslational processing, including glycosylation and phosphorylation. The molecular weight (MW) of fully modified fetuin-A (FM-fetuin-A) is approximately 60 kDa in an immunoblot, which is much higher than the estimated MW by amino acid sequence. Under conditions of calcification stress such as advanced stage chronic kidney disease, fetuin-A prevents calcification by forming colloidal complexes, which are referred to as calciprotein particles (CPP). Since the significance of CPP in this process is unclear, we investigated the effect of synthetic secondary CPP on the level of FM-fetuin-A in HepG2 cells. Secondary CPP increased the level of FM-fetuin-A in dose- and time-dependent manners, but did not affect expression of mRNA for fetuin-A. Treatment with O- and/or N-glycosidase caused a shift of the 60 kDa band of FM-fetuin-A to a lower MW. Preincubation with brefeldin A, an inhibitor of transport of newly synthesized proteins from the endoplasmic reticulum to the Golgi apparatus, completely blocked the secondary CPP-induced increase in FM-fetuin-A. Treatment with BAPTA-AM, an intracellular calcium chelating agent, also inhibited the CPP-induced increase in the FM-fetuin-A level. Secondary CPP accelerate posttranslational processing of fetuin-A in HepG2 cells.
Collapse
Affiliation(s)
- Hideki Uedono
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Katsuhito Mori
- Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - Akinobu Ochi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuya Miki
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Tsuda
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuki Nagata
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuo Imanishi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaaki Inaba
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masanori Emoto
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
26
|
Choo P, Liu T, Odom TW. Nanoparticle Shape Determines Dynamics of Targeting Nanoconstructs on Cell Membranes. J Am Chem Soc 2021; 143:4550-4555. [PMID: 33735562 DOI: 10.1021/jacs.1c00850] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanoparticle carriers are effective drug delivery vehicles. Along with other design parameters including size, composition, and surface charge, particle shape strongly influences cellular uptake. How nanoparticle geometry affects targeted delivery under physiologically relevant conditions, however, is inconclusive. Here, we demonstrate that nanoconstruct core shape influences the dynamics of targeting ligand-receptor interactions on cancer cell membranes. By single-particle tracking of translational and rotational motion, we compared DNA aptamer AS1411 conjugated gold nanostars (AS1411-AuNS) and 50 nm gold spheres (AS1411-50NPs) on cells with and without targeted nucleolin membrane receptors. On nucleolin-expressing cells, AS1411-AuNS exhibited faster velocities under directed diffusion and translated over larger areas during restricted diffusion compared to AS1411-50NPs, despite their similar protein corona profiles. On nucleolin-inhibited cells, AS1411-AuNS showed faster rotation dynamics over smaller translational areas, while AS1411-50NPs did not display significant changes in translation. These differences in translational and rotational motions indicate that nanoparticle shape affects how targeting nanoconstructs bind to cell-membrane receptors.
Collapse
|
27
|
Kuro-O M. Phosphate as a Pathogen of Arteriosclerosis and Aging. J Atheroscler Thromb 2021; 28:203-213. [PMID: 33028781 PMCID: PMC8048948 DOI: 10.5551/jat.rv17045] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
During the evolution of skeletons, terrestrial vertebrates acquired strong bones made of calcium-phosphate. By keeping the extracellular fluid in a supersaturated condition regarding calcium and phosphate ions, they created the bone when and where they wanted simply by providing a cue for precipitation. To secure this strategy, they acquired a novel endocrine system to strictly control the extracellular phosphate concentration. In response to phosphate intake, fibroblast growth factor-23 (FGF23) is secreted from the bone and acts on the kidney through binding to its receptor Klotho to increase urinary phosphate excretion, thereby maintaining phosphate homeostasis. The FGF23-Klotho endocrine system, when disrupted in mice, results in hyperphosphatemia and vascular calcification. Besides, mice lacking Klotho or FGF23 suffer from complex aging-like phenotypes, which are alleviated by placing them on a low- phosphate diet, indicating that phosphate is primarily responsible for the accelerated aging. Phosphate acquires the ability to induce cell damage and inflammation when precipitated with calcium. In the blood, calcium-phosphate crystals are adsorbed by serum protein fetuin-A and prevented from growing into large precipitates. Consequently, nanoparticles that comprised calcium-phosphate crystals and fetuin-A, termed calciprotein particles (CPPs), are generated and dispersed as colloids. CPPs increase in the blood with an increase in serum phosphate and age. Circulating CPP levels correlate positively with vascular stiffness and chronic non-infectious inflammation, raising the possibility that CPPs may be an endogenous pro-aging factor. Terrestrial vertebrates with the bone made of calcium- phosphate may be destined to age due to calcium-phosphate in the blood.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Calciprotein particles (CPP) are formed in supersaturated solutions of calcium, phosphate and the mineral-binding protein fetuin-A. CPP have garnered considerable interest as potential mediators of mineral stress, but little consideration has been given to their origin, clearance and role in metabolism. RECENT FINDINGS CPP are made whilst buffering the mineral absorbed from the intestine after a meal or during remodelling of bone matrix. The postprandial rise in circulating CPP rise may be sensed by osteoblasts/osteocytes in bone, stimulating the secretion of the master phosphatonin fibroblast growth factor 23. Amorphous calcium phosphate-containing CPP are rapidly cleared by endothelial cells in the liver whereas crystalline apatite-containing CPP are filtered by phagocytic cells of the reticuloendothelial system. Impaired excretory function in kidney disease may lead to accumulation of CPP and its precursors with possible pathological sequalae. Inability to stabilize CPP in fetuin-A-deficiency states can result in intraluminal precipitation and inflammatory cascades if other mineralisation regulatory networks are compromised. SUMMARY CPP allow efficient transport and clearance of bulk calcium phosphate as colloids without risk of precipitation. As circulating factors, CPP may couple dietary mineral exposure with endocrine control of mineral metabolism in bone, signalling the need to dispose of excess phosphate from the body.
Collapse
|
29
|
Rudloff S, Janot M, Rodriguez S, Dessalle K, Jahnen-Dechent W, Huynh-Do U. Fetuin-A is a HIF target that safeguards tissue integrity during hypoxic stress. Nat Commun 2021; 12:549. [PMID: 33483479 PMCID: PMC7822914 DOI: 10.1038/s41467-020-20832-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 12/18/2020] [Indexed: 02/08/2023] Open
Abstract
Intrauterine growth restriction (IUGR) is associated with reduced kidney size at birth, accelerated renal function decline, and increased risk for chronic kidney and cardiovascular diseases in adults. Precise mechanisms underlying fetal programming of adult diseases remain largely elusive and warrant extensive investigation. Setting up a mouse model of hypoxia-induced IUGR, fetal adaptations at mRNA, protein and cellular levels, and their long-term functional consequences are characterized, using the kidney as a readout. Here, we identify fetuin-A as an evolutionary conserved HIF target gene, and further investigate its role using fetuin-A KO animals and an adult model of ischemia-reperfusion injury. Beyond its role as systemic calcification inhibitor, fetuin-A emerges as a multifaceted protective factor that locally counteracts calcification, modulates macrophage polarization, and attenuates inflammation and fibrosis, thus preserving kidney function. Our study paves the way to therapeutic approaches mitigating mineral stress-induced inflammation and damage, principally applicable to all soft tissues.
Collapse
Affiliation(s)
- Stefan Rudloff
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Mathilde Janot
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Stephane Rodriguez
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Onco-haematology, Geneva Medical University, Geneva, Switzerland
| | - Kevin Dessalle
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Medical Faculty, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland.
| |
Collapse
|
30
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
31
|
Urinary phosphate-containing nanoparticle contributes to inflammation and kidney injury in a salt-sensitive hypertension rat model. Commun Biol 2020; 3:575. [PMID: 33060834 PMCID: PMC7562875 DOI: 10.1038/s42003-020-01298-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Although disturbed phosphate metabolism frequently accompanies chronic kidney disease (CKD), its causal role in CKD progression remains unclear. It is also not fully understood how excess salt induces organ damage. We here show that urinary phosphate-containing nanoparticles promote kidney injury in salt-sensitive hypertension. In Dahl salt-sensitive rats, salt loading resulted in a significant increase in urinary phosphate excretion without altering serum phosphate levels. An intestinal phosphate binder sucroferric oxyhydroxide attenuated renal inflammation and proteinuria in this model, along with the suppression of phosphaturia. Using cultured proximal tubule cells, we confirmed direct pathogenic roles of phosphate-containing nanoparticles in renal tubules. Finally, transcriptome analysis revealed a potential role of complement C1q in renal inflammation associated with altered phosphate metabolism. These data demonstrate that increased phosphate excretion promotes renal inflammation in salt-sensitive hypertension and suggest a role of disturbed phosphate metabolism in the pathophysiology of hypertensive kidney disease and high salt-induced kidney injury.
Collapse
|
32
|
Carbonate Apatite and Hydroxyapatite Formulated with Minimal Ingredients to Deliver SiRNA into Breast Cancer Cells In Vitro and In Vivo. J Funct Biomater 2020; 11:jfb11030063. [PMID: 32927738 PMCID: PMC7565062 DOI: 10.3390/jfb11030063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/24/2020] [Accepted: 09/07/2020] [Indexed: 01/22/2023] Open
Abstract
Introduction: Cancer is one of the top-ranked noncommunicable diseases causing deaths to nine million people and affecting almost double worldwide in 2018. Tremendous advancement in surgery, chemotherapy, radiation and targeted immunotherapy have improved the rate of cure and disease-free survival. As genetic mutations vary in different cancers, potential of customized treatment to silence the problem gene/s at the translational level is being explored too. Yet delivering therapeutics at the required dosage only to the affected cells without affecting the healthy ones, is a big hurdle to be overcome. Scientists worldwide have been working to invent a smart drug delivery system for targeted delivery of therapeutics to tumor tissues only. As part of such an effort, few organic nanocarriers went to clinical trials, while inorganic nanoparticles (NPs) are still in development stage despite their many customizable properties. Carbonate apatite (CA), a pH sensitive nanocarrier has emerged as an efficient delivery system for drugs, plasmids and siRNAs in preclinical models of breast and colon cancers. Like hydroxyapatite (HA) which serves as a classical tool for delivery of genetic materials such as siRNA and plasmid, CA is an apatite-based synthetic carrier. We developed simplified methods of formulating CA-in-DMEM and a DMEM-mimicking buffer and HA in a HEPES-buffered solution and characterized them in terms of size, stability, protein corona (PC) composition, cytotoxicity, siRNA delivery efficiency in breast cancer cells and siRNA biodistribution profile in a mouse model of breast cancer. Methods: Particle growth was analyzed via spectrophotometry and light microscopy, size was measured via dynamic light scattering and scanning electron microscopy and confirmation of functional groups in apatite structures was made by FT-IR. siRNA-binding was analyzed via spectrophotometry. Stability of the formulation solutions/buffers was tested over various time points and at different temperatures to determine their compatibility in the context of practical usage. Cellular uptake was studied via fluorescence microscopy. MTT assay was performed to measure the cytotoxicity of the NPs. Liquid chromatography—mass spectrometry was carried out to analyze the PC formed around all three different NPs in serum-containing media. To explore biodistribution of all the formulations, fluorescence-labeled siRNA-loaded NPs were administered intravenously prior to analysis of fluorescence intensity in the collected organs and tumors of the treated mice. Results: The size of NPs in 10% serum-containing media was dramatically different where CA-in-DMB and HA were much larger than CA-in-DMEM. Effect of media was notable on the PC composition of all three NPs. All three NPs bound albumin and some common protease inhibitors involved in bone metabolism due to their compositional similarity to our bone materials. Moreover, CA also bound heme-binding proteins and opsonins. Unlike CA, HA bound different kinds of keratins. Difference in PC constitution was likely to influence accumulation of NPs in various organs including those of reticuloendothelial system, such as liver and spleen and the tumor. We found 10 times more tumor accumulation of CA-in-DMB than CA-in-DMEM, which could be due to more stable siRNA-binding and distinct PC composition of the former. Conclusion: As a nanocarrier CA is more efficient than HA for siRNA delivery to the tumor. CA prepared in a buffer containing only the mere constituents was potentially more efficient than classical CA prepared in DMEM, owing to the exclusion of interference attributed by the inorganic ions and organic molecules present in DMEM.
Collapse
|
33
|
Fan HX, Gou BD, Gao YX, Wu G, Liu SH, Li F, Zhang TL. Protein-bound calcium phosphate in uremic rat serum: a quantitative study. J Biol Inorg Chem 2020; 25:1051-1063. [PMID: 32839886 DOI: 10.1007/s00775-020-01807-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/04/2020] [Indexed: 10/23/2022]
Abstract
Protein-bound calcium (prCa) constitutes about 40% of serum total calcium, in which albumin is the most dominant protein. Given the chemical interaction between calcium and phosphate (Pi), the increased serum Pi in chronic kidney disease may cause changes in the composition and structure of the prCa fraction. Here, we report the phosphate binding on the protein-bound calcium in uremic rat serum. Using adenine-fed rats as a uremic model, we separated the calcium and phosphate fractions in rat serum by ultrafiltration, and found that the level of protein-bound phosphate (prPi) in the uremic serum was markedly higher than in control. The elevated prPi level was comparable to the prCa level, consistent with the presence of protein-bound calcium phosphate pr(Ca)j-m(CaPi)m. We then confirmed its presence by ex vivo X-ray absorption near-edge structure spectroscopy, revealing the discrete state of the calcium phosphate clusters associated with protein. Finally, in a quantitative investigation using Ca- and Pi-boosted serum, we discovered the threshold concentration for the Pi binding on prCa, and determined the binding constant. The threshold, while preventing Pi from binding to prCa in normal condition, allows the reaction to take place in hyperphosphatemia conditions. The protein-bound calcium phosphate could act as a link between the metabolism of serum proteins and the homeostasis of phosphate and calcium, and it deserves further investigation whether the molar ratio of (prPi/prCa)⋅100% may serve as a serum index of the vascular calcification status in chronic kidney disease.
Collapse
Affiliation(s)
- Hong-Xing Fan
- Department of Preclinical and Forensic Medicine, Baotou Medical College, Inner Mongolia University of Science & Technology, Baotou, 014010, People's Republic of China.,CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, 19B Yuquan Road, Beijing, 100049, People's Republic of China
| | - Bao-Di Gou
- Department of Chemical Biology, Peking University School of Pharmaceutical Sciences, 38 Xueyuan Road, Beijing, 100191, People's Republic of China
| | - Yu-Xi Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, 19B Yuquan Road, Beijing, 100049, People's Republic of China
| | - Gang Wu
- Department of Preclinical and Forensic Medicine, Baotou Medical College, Inner Mongolia University of Science & Technology, Baotou, 014010, People's Republic of China.
| | - Shu-Hu Liu
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, 19B Yuquan Road, Beijing, 100049, People's Republic of China
| | - Fan Li
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, 19B Yuquan Road, Beijing, 100049, People's Republic of China
| | - Tian-Lan Zhang
- Department of Chemical Biology, Peking University School of Pharmaceutical Sciences, 38 Xueyuan Road, Beijing, 100191, People's Republic of China.
| |
Collapse
|
34
|
Chen W, Fitzpatrick J, Monroy-Trujillo JM, Sozio SM, Jaar BG, Estrella MM, Serrano J, Anokhina V, Miller BL, Melamed ML, Bushinsky DA, Parekh RS. Associations of Serum Calciprotein Particle Size and Transformation Time With Arterial Calcification, Arterial Stiffness, and Mortality in Incident Hemodialysis Patients. Am J Kidney Dis 2020; 77:346-354. [PMID: 32800846 DOI: 10.1053/j.ajkd.2020.05.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/20/2020] [Indexed: 01/09/2023]
Abstract
RATIONALE & OBJECTIVE Characteristics of the transformation of primary to secondary calciprotein particles (CPPs) in serum, including the size of secondary CPP (CPP2) aggregates and the time of transformation (T50), may be markers for arterial calcification in patients undergoing hemodialysis (HD). We examined the associations of CPP2 aggregate size and T50 with arterial calcification in incident HD patients. STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS Incident HD patients (n=402with available CPP2 measures and n=388with available T50 measures) from the Predictors of Arrhythmic and Cardiovascular Risk in End-Stage Renal Disease (PACE) Study PREDICTORS: Serum CPP2 size and T50 at baseline. OUTCOMES Primary outcomes were baseline coronary artery and thoracic aorta calcifications. Exploratory outcomes included baseline arterial stiffness, measured by pulse wave velocity (PWV) and ankle brachial index, and longitudinally, repeat measures of PWV and all-cause mortality. ANALYTICAL APPROACH Tobit regression, multiple linear regression, Poisson regression, linear mixed-effects regression, and Cox proportional hazards regression. RESULTS Mean age was 55±13 years, 41% were women, 71% were Black, and 57% had diabetes mellitus. Baseline CPP2 size and T50 were correlated with baseline fetuin A level (r=-0.59 for CPP2 and 0.44 for T50; P<0.001 for both), but neither was associated with baseline measures of arterial calcification or arterial stiffness. Baseline CPP2 size and T50 were not associated with repeat measures of PWV. During a median follow-up of 3.5 (IQR, 1.7-6.2) years, larger CPP2 was associated with higher risk for mortality (HR, 1.17 [95% CI, 1.05-1.31] per 100nm larger CPP2 size) after adjusting for demographics and comorbid conditions, but there was no association between baseline T50 and risk for mortality. LIMITATIONS Possible imprecision in assays, small sample size, limited generalizability to incident HD populations with different racial composition, and residual confounding. CONCLUSIONS In incident HD patients, neither CPP2 size nor T50 was associated with prevalent arterial calcification and stiffness. Larger CPP2 was associated with risk for mortality, but this finding needs to be confirmed in future studies.
Collapse
Affiliation(s)
- Wei Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| | - Jessica Fitzpatrick
- Department of Medicine and Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | | | - Stephen M Sozio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD
| | - Bernard G Jaar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD; Nephrology Center of Maryland, Baltimore, MD
| | - Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine, University of California, San Francisco, CA; San Francisco VA Health Care System, San Francisco, CA
| | - Jishyra Serrano
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Viktoriya Anokhina
- Departments of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Benjamin L Miller
- Departments of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY; Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY; Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Michal L Melamed
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - David A Bushinsky
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Rulan S Parekh
- Department of Medicine and Pediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
35
|
Jahnen-Dechent W, Büscher A, Köppert S, Heiss A, Kuro-O M, Smith ER. Mud in the blood: the role of protein-mineral complexes and extracellular vesicles in biomineralisation and calcification. J Struct Biol 2020; 212:107577. [PMID: 32711043 DOI: 10.1016/j.jsb.2020.107577] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022]
Abstract
Protein-mineral interaction is known to regulate biomineral stability and morphology. We hypothesise that fluid phases produce highly dynamic protein-mineral complexes involved in physiology and pathology of biomineralisation. Here, we specifically focus on calciprotein particles, complexes of vertebrate mineral-binding proteins and calcium phosphate present in the systemic circulation and abundant in extracellular fluids - hence the designation of the ensuing protein-mineral complexes as "mud in the blood". These complexes exist amongst other extracellular particles that we collectively refer to as "the particle zoo".
Collapse
Affiliation(s)
- Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany.
| | - Andrea Büscher
- Helmholtz-Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Sina Köppert
- Helmholtz-Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Alexander Heiss
- The Research Institute for Precious Metals and Metals Chemistry (fem), Schwaebisch Gmuend, Germany
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Medicine, University of Melbourne, Parkville, Australia
| |
Collapse
|
36
|
Candellier A, Hénaut L, Morelle J, Choukroun G, Jadoul M, Brazier M, Goffin É. Aortic stenosis in patients with kidney failure: Is there an advantage for a PD-first policy? Perit Dial Int 2020; 41:158-167. [DOI: 10.1177/0896860820941371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Aortic stenosis (AS) is the most common valvular disease. It is twice as prevalent in patients with kidney failure as compared to the general population. In addition, AS progresses at a faster rate and is associated with a higher risk of death and poorer quality of life in patients on dialysis. Chronic kidney disease–mineral and bone disorder (CKD-MBD), inflammation, and hemodynamic disturbances contribute to the pathophysiology and progression of AS. Whether the type of dialysis modality, that is, hemodialysis (HD) versus peritoneal dialysis (PD), has a differential impact on the development and progression of AS in patients with kidney failure remains debated. Recent data indicate that the prevalence of valvular calcifications might be lower and the development of AS delayed in PD patients, as compared to those treated with HD. This could be accounted for by several mechanisms including reduced valvular shear stress, better preservation of residual kidney function (with better removal of protein-bound uremic toxins and CKD-MBD profile), and lower levels of systemic inflammation. Given the high morbidity and mortality rates related to interventional procedures in the population with kidney failure, surgical and transcatheter aortic valve replacement should be considered in selected patients with severe AS. Strategies slowing down the progression of aortic valve remodeling should remain the cornerstone in the management of individuals with kidney failure and mild to moderate AS. This review explores the potential benefits of PD in patients with kidney failure and AS and provides some clues to help clinicians in the decision-making process when options for kidney replacement therapy are considered in patients with AS.
Collapse
Affiliation(s)
- Alexandre Candellier
- Division of Nephrology, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR UPJV 7517, MP3CV, CURS, Amiens, France
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Johann Morelle
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Gabriel Choukroun
- Division of Nephrology, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR UPJV 7517, MP3CV, CURS, Amiens, France
| | - Michel Jadoul
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | | | - Éric Goffin
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| |
Collapse
|
37
|
Calciprotein Particles and Serum Calcification Propensity: Hallmarks of Vascular Calcifications in Patients with Chronic Kidney Disease. J Clin Med 2020; 9:jcm9051287. [PMID: 32365608 PMCID: PMC7288330 DOI: 10.3390/jcm9051287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular complications are one of the leading causes of mortality worldwide and are strongly associated with atherosclerosis and vascular calcification (VC). Patients with chronic kidney disease (CKD) have a higher prevalence of VC as renal function declines, which will result in increased mortality. Serum calciprotein particles (CPPs) are colloidal nanoparticles that have a prominent role in the initiation and progression of VC. The T50 test is a novel test that measures the conversion of primary to secondary calciprotein particles indicating the tendency of serum to calcify. Therefore, we accomplished a comprehensive review as the first integrated approach to clarify fundamental aspects that influence serum CPP levels and T50, and to explore the effects of CPP and calcification propensity on various chronic disease outcomes. In addition, new topics were raised regarding possible clinical uses of T50 in the assessment of VC, particularly in patients with CKD, including possible opportunities in VC management. The relationships between serum calcification propensity and cardiovascular and all-cause mortality were also addressed. The review is the outcome of a comprehensive search on available literature and could open new directions to control VC.
Collapse
|
38
|
Yuan C, Aierken A, Xie Z, Li N, Zhao J, Qing H. The age-related microglial transformation in Alzheimer's disease pathogenesis. Neurobiol Aging 2020; 92:82-91. [PMID: 32408056 DOI: 10.1016/j.neurobiolaging.2020.03.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/11/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022]
Abstract
Neuroinflammatory responses mediated by microglia, the resident immune cells of the central nervous system, have long been a subject of study in the field of Alzheimer's disease (AD). Microglia express a wide range of receptors that act as molecular sensors, through which they can fulfill their various functions. In this review, we first analyzed the changes in the expression levels of microglial membrane receptors SR-A, TREM2, CD36, CD33, and CR3 in aging and AD and described the different roles of these receptors in amyloid-beta clearance and inflammatory responses. Two classical hallmarks of AD are extracellular amyloid-beta deposits and intracellular aggregated phosphorylated tau. In AD, microglia reaction was initially thought to be triggered by amyloid deposits. New evidence showed it also associated with increased phosphorylation of tau. However, which first appeared and induced activated microglia is not clear. Then we summarized diverse opinions on it. Besides, as AD is tightly linked to aging, and microglia changes dramatically on aging, yet the relative impacts of both aging and microglia are less frequently considered, so at last, we discussed the roles of aging microglia in AD. We hope to provide a reference for subsequent research.
Collapse
Affiliation(s)
- Chunxu Yuan
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ailikemu Aierken
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Nuomin Li
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Juan Zhao
- School of Materials Science and Engineering, Department of Materials Processing Engineering, Beijing Institute of Technology, Beijing, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
39
|
Chen W, Anokhina V, Dieudonne G, Abramowitz MK, Kashyap R, Yan C, Wu TT, de Mesy Bentley KL, Miller BL, Bushinsky DA. Patients with advanced chronic kidney disease and vascular calcification have a large hydrodynamic radius of secondary calciprotein particles. Nephrol Dial Transplant 2020; 34:992-1000. [PMID: 29788425 DOI: 10.1093/ndt/gfy117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The size of secondary calciprotein particles (CPP2) and the speed of transformation (T50) from primary calciprotein particles (CPP1) to CPP2 in serum may be associated with vascular calcification (VC) in patients with chronic kidney disease (CKD). METHODS We developed a high throughput, microplate-based assay using dynamic light scattering (DLS) to measure the transformation of CPP1 to CPP2, hydrodynamic radius (Rh) of CPP1 and CPP2, T50 and aggregation of CPP2. We used this DLS assay to test the hypothesis that a large Rh of CPP2 and/or a fast T50 are associated with VC in 45 participants with CKD Stages 4-5 (22 without VC and 23 with VC) and 17 healthy volunteers (HV). VC was defined as a Kauppila score >6 or an Adragao score ≥3. RESULTS CKD participants with VC had larger cumulants Rh of CPP2 {370 nm [interquartile range (IQR) 272-566]} compared with CKD participants without VC [212 nm (IQR 169-315)] and compared with HV [168 nm (IQR 145-352), P < 0.01 for each]. More CPP2 were in aggregates in CKD participants with VC than those without VC (70% versus 36%). The odds of having VC increased by 9% with every 10 nm increase in the Rh of CPP2, after adjusting for age, diabetes, serum calcium and phosphate [odds ratio 1.09, 95% confidence interval (CI) 1.03, 1.16, P = 0.005]. The area under the receiver operating characteristic curve for VC of CPP2 size was 0.75 (95% CI 0.60, 0.90). T50 was similar in CKD participants with and without VC, although both groups had a lower T50 than HV. CONCLUSIONS Rh of CPP2, but not T50, is independently associated with VC in patients with CKD Stages 4-5.
Collapse
Affiliation(s)
- Wei Chen
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Viktoriya Anokhina
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Gregory Dieudonne
- Department of Radiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Randeep Kashyap
- Department of Surgery, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Chen Yan
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Tong Tong Wu
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Karen L de Mesy Bentley
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Benjamin L Miller
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - David A Bushinsky
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
40
|
Magnesium and calciprotein particles in vascular calcification: the good cop and the bad cop. Curr Opin Nephrol Hypertens 2020; 28:368-374. [PMID: 31045659 DOI: 10.1097/mnh.0000000000000509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Vascular calcification is a major contributor to increased cardiovascular mortality in chronic kidney disease (CKD). Recently, calciprotein particles (CPP) were identified to drive the calcification process. CPP may explain the effects of high phosphate on vascular calcification. Magnesium is a promising novel therapeutic approach to halt vascular calcification, because it inhibits CPP maturation and is associated with reduced cardiovascular mortality in CKD. We aim to examine the current evidence for the role of CPP in the calcification process and to explain how magnesium prevents calcification. RECENT FINDINGS A recent meta-analysis concluded that reducing high phosphate levels in CKD patients does not associate with lowering cardiovascular mortality. Inhibition of CPP formation prevents phosphate-induced calcification in vitro. Consequently, delaying CPP formation and maturation may be a clinical approach to reduce calcification. Magnesium inhibits CPP maturation and vascular calcification. Clinical pilot studies suggest that magnesium is a promising intervention strategy against calcification in CKD patients. SUMMARY CPP induce vascular calcification and are modulated by serum phosphate and magnesium concentrations. Magnesium is a strong inhibitor of CPP maturation and therefore, a promising therapeutic approach to reduce vascular calcification in CKD. Currently, several studies are being performed to determine the clinical outcomes of magnesium supplementation in CKD.
Collapse
|
41
|
Hou YC, Lu CL, Zheng CM, Liu WC, Yen TH, Chen RM, Lin YF, Chao CT, Lu KC. The Role of Vitamin D in Modulating Mesenchymal Stem Cells and Endothelial Progenitor Cells for Vascular Calcification. Int J Mol Sci 2020; 21:ijms21072466. [PMID: 32252330 PMCID: PMC7177675 DOI: 10.3390/ijms21072466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Vascular calcification, which involves the deposition of calcifying particles within the arterial wall, is mediated by atherosclerosis, vascular smooth muscle cell osteoblastic changes, adventitial mesenchymal stem cell osteoblastic differentiation, and insufficiency of the calcification inhibitors. Recent observations implied a role for mesenchymal stem cells and endothelial progenitor cells in vascular calcification. Mesenchymal stem cells reside in the bone marrow and the adventitial layer of arteries. Endothelial progenitor cells that originate from the bone marrow are an important mechanism for repairing injured endothelial cells. Mesenchymal stem cells may differentiate osteogenically by inflammation or by specific stimuli, which can activate calcification. However, the bioactive substances secreted from mesenchymal stem cells have been shown to mitigate vascular calcification by suppressing inflammation, bone morphogenetic protein 2, and the Wingless-INT signal. Vitamin D deficiency may contribute to vascular calcification. Vitamin D supplement has been used to modulate the osteoblastic differentiation of mesenchymal stem cells and to lessen vascular injury by stimulating adhesion and migration of endothelial progenitor cells. This narrative review clarifies the role of mesenchymal stem cells and the possible role of vitamin D in the mechanisms of vascular calcification.
Collapse
Affiliation(s)
- Yi-Chou Hou
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, New Taipei City 231, Taiwan;
- School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
| | - Chien-Lin Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan;
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City 243, Taiwan
| | - Cai-Mei Zheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
| | - Wen-Chih Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, Tungs’ Taichung Metroharbor Hospital, Taichung City 43304, Taiwan
| | - Tzung-Hai Yen
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
| | - Chia-Ter Chao
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 104, Taiwan
- Nephrology division, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 108, Taiwan
- Correspondence: (C.-T.C.); (K.-C.L.)
| | - Kuo-Cheng Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan;
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City 243, Taiwan
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan
- Correspondence: (C.-T.C.); (K.-C.L.)
| |
Collapse
|
42
|
Akiyama KI, Miura Y, Hayashi H, Sakata A, Matsumura Y, Kojima M, Tsuchiya K, Nitta K, Shiizaki K, Kurosu H, Kuro-o M. Calciprotein particles regulate fibroblast growth factor-23 expression in osteoblasts. Kidney Int 2020; 97:702-712. [DOI: 10.1016/j.kint.2019.10.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 09/24/2019] [Accepted: 10/10/2019] [Indexed: 11/26/2022]
|
43
|
Herrmann M, Babler A, Moshkova I, Gremse F, Kiessling F, Kusebauch U, Nelea V, Kramann R, Moritz RL, McKee MD, Jahnen-Dechent W. Lumenal calcification and microvasculopathy in fetuin-A-deficient mice lead to multiple organ morbidity. PLoS One 2020; 15:e0228503. [PMID: 32074120 PMCID: PMC7029858 DOI: 10.1371/journal.pone.0228503] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/16/2020] [Indexed: 12/22/2022] Open
Abstract
The plasma protein fetuin-A mediates the formation of protein-mineral colloids known as calciprotein particles (CPP)-rapid clearance of these CPP by the reticuloendothelial system prevents errant mineral precipitation and therefore pathological mineralization (calcification). The mutant mouse strain D2,Ahsg-/- combines fetuin-A deficiency with the calcification-prone DBA/2 genetic background, having a particularly severe compound phenotype of microvascular and soft tissue calcification. Here we studied mechanisms leading to soft tissue calcification, organ damage and death in these mice. We analyzed mice longitudinally by echocardiography, X-ray-computed tomography, analytical electron microscopy, histology, mass spectrometry proteomics, and genome-wide microarray-based expression analyses of D2 wildtype and Ahsg-/- mice. Fetuin-A-deficient mice had calcified lesions in myocardium, lung, brown adipose tissue, reproductive organs, spleen, pancreas, kidney and the skin, associated with reduced growth, cardiac output and premature death. Importantly, early-stage calcified lesions presented in the lumen of the microvasculature suggesting precipitation of mineral containing complexes from the fluid phase of blood. Genome-wide expression analysis of calcified lesions and surrounding (not calcified) tissue, together with morphological observations, indicated that the calcification was not associated with osteochondrogenic cell differentiation, but rather with thrombosis and fibrosis. Collectively, these results demonstrate that soft tissue calcification can start by intravascular mineral deposition causing microvasculopathy, which impacts on growth, organ function and survival. Our study underscores the importance of fetuin-A and related systemic regulators of calcified matrix metabolism to prevent cardiovascular disease, especially in dysregulated mineral homeostasis.
Collapse
Affiliation(s)
- Marietta Herrmann
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Anne Babler
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Irina Moshkova
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Felix Gremse
- Helmholtz Institute for Biomedical Engineering, Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Fabian Kiessling
- Helmholtz Institute for Biomedical Engineering, Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Ulrike Kusebauch
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Valentin Nelea
- Faculty of Dentistry, Faculty of Medicine (Dept. of Anatomy and Cell Biology), McGill University, Montreal, Quebec, Canada
| | - Rafael Kramann
- Division of Nephrology, RWTH Aachen University Hospital, Aachen, Germany
| | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Marc D. McKee
- Faculty of Dentistry, Faculty of Medicine (Dept. of Anatomy and Cell Biology), McGill University, Montreal, Quebec, Canada
| | - Willi Jahnen-Dechent
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
44
|
Babler A, Schmitz C, Buescher A, Herrmann M, Gremse F, Gorgels T, Floege J, Jahnen-Dechent W. Microvasculopathy and soft tissue calcification in mice are governed by fetuin-A, magnesium and pyrophosphate. PLoS One 2020; 15:e0228938. [PMID: 32074140 PMCID: PMC7029863 DOI: 10.1371/journal.pone.0228938] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/27/2020] [Indexed: 12/18/2022] Open
Abstract
Calcifications can disrupt organ function in the cardiovascular system and the kidney, and are particularly common in patients with chronic kidney disease (CKD). Fetuin-A deficient mice maintained against the genetic background DBA/2 exhibit particularly severe soft tissue calcifications, while fetuin-A deficient C57BL/6 mice remain healthy. We employed molecular genetic analysis to identify risk factors of calcification in fetuin-A deficient mice. We sought to identify pharmaceutical therapeutic targets that could be influenced by dietary of parenteral supplementation. We studied the progeny of an intercross of fetuin-A deficient DBA/2 and C57BL/6 mice to identify candidate risk genes involved in calcification. We determined that a hypomorphic mutation of the Abcc6 gene, a liver ATP transporter supplying systemic pyrophosphate, and failure to regulate the Trpm6 magnesium transporter in kidney were associated with severity of calcification. Calcification prone fetuin-A deficient mice were alternatively treated with parenteral administration of fetuin-A dietary magnesium supplementation, phosphate restriction, or by or parenteral pyrophosphate. All treatments markedly reduced soft tissue calcification, demonstrated by computed tomography, histology and tissue calcium measurement. We show that pathological ectopic calcification in fetuin-A deficient DBA/2 mice is caused by a compound deficiency of three major extracellular and systemic inhibitors of calcification, namely fetuin-A, magnesium, and pyrophosphate. All three of these are individually known to contribute to stabilize protein-mineral complexes and thus inhibit mineral precipitation from extracellular fluid. We show for the first time a compound triple deficiency that can be treated by simple dietary or parenteral supplementation. This is of special importance in patients with advanced CKD, who commonly exhibit reduced serum fetuin-A, magnesium and pyrophosphate levels.
Collapse
Affiliation(s)
- Anne Babler
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Carlo Schmitz
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Andrea Buescher
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| | - Marietta Herrmann
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
- IZKF Research Group Tissue Regeneration in Musculoskeletal Regeneration, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Felix Gremse
- Helmholtz Institute for Biomedical Engineering, Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Theo Gorgels
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Juergen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Willi Jahnen-Dechent
- Helmholtz Institute for Biomedical Engineering, Biointerface Lab, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
45
|
Schantl AE, Verhulst A, Neven E, Behets GJ, D'Haese PC, Maillard M, Mordasini D, Phan O, Burnier M, Spaggiari D, Decosterd LA, MacAskill MG, Alcaide-Corral CJ, Tavares AAS, Newby DE, Beindl VC, Maj R, Labarre A, Hegde C, Castagner B, Ivarsson ME, Leroux JC. Inhibition of vascular calcification by inositol phosphates derivatized with ethylene glycol oligomers. Nat Commun 2020; 11:721. [PMID: 32024848 PMCID: PMC7002685 DOI: 10.1038/s41467-019-14091-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022] Open
Abstract
Myo-inositol hexakisphosphate (IP6) is a natural product known to inhibit vascular calcification (VC), but with limited potency and low plasma exposure following bolus administration. Here we report the design of a series of inositol phosphate analogs as crystallization inhibitors, among which 4,6-di-O-(methoxy-diethyleneglycol)-myo-inositol-1,2,3,5-tetrakis(phosphate), (OEG2)2-IP4, displays increased in vitro activity, as well as more favorable pharmacokinetic and safety profiles than IP6 after subcutaneous injection. (OEG2)2-IP4 potently stabilizes calciprotein particle (CPP) growth, consistently demonstrates low micromolar activity in different in vitro models of VC (i.e., human serum, primary cell cultures, and tissue explants), and largely abolishes the development of VC in rodent models, while not causing toxicity related to serum calcium chelation. The data suggest a mechanism of action independent of the etiology of VC, whereby (OEG2)2-IP4 disrupts the nucleation and growth of pathological calcification.
Collapse
Affiliation(s)
- Antonia E Schantl
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Anja Verhulst
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Ellen Neven
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Geert J Behets
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Marc Maillard
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - David Mordasini
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Phan
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Burnier
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - Dany Spaggiari
- Division of Clinical Pharmacology, Lausanne University Hospital, Lausanne, Switzerland
| | - Laurent A Decosterd
- Division of Clinical Pharmacology, Lausanne University Hospital, Lausanne, Switzerland
| | - Mark G MacAskill
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Carlos J Alcaide-Corral
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Adriana A S Tavares
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Victoria C Beindl
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | - Anne Labarre
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Chrismita Hegde
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Bastien Castagner
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | | | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
46
|
Clemente A, Traghella I, Mazzone A, Sbrana S, Vassalle C. Vascular and valvular calcification biomarkers. Adv Clin Chem 2020; 95:73-103. [PMID: 32122525 DOI: 10.1016/bs.acc.2019.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vascular and valvular calcification constitutes a major health problem with serious clinical consequences. It is important for medical laboratorians to improve their knowledge on this topic and to know which biological markers may have a potential interest and might be useful for diagnosis and for management of ectopic calcifications. This review focuses on the pathophysiological mechanisms of vascular and valvular calcification, with emphasis on the mechanisms that are different for the two types of events, which underscore the need for differentiated healthcare, and explain different response to therapy. Available imaging and scoring tools used to assess both vascular and valvular calcification, together with the more studied and reliable biological markers emerging in this field (e.g., Fetuin A and matrix Gla protein), are discussed. Recently proposed functional assays, measuring the propensity of human serum to calcify, appear promising for vascular calcification assessment and are described. Further advancement through omic technologies and statistical tools is also reported. Clinical chemistry and laboratory medicine practitioners overlook this new era that will engage them in the near future, where a close cooperation of professionals with different competencies, including laboratorists, is required. This innovative approach may truly revolutionize practice of laboratory and of whole medicine attitude, making progression in knowledge of pathways relevant to health, as the complex calcification-related pathways, and adding value to patient care, through a precision medicine strategy.
Collapse
|
47
|
Vidaud C, Miccoli L, Brulfert F, Aupiais J. Fetuin exhibits a strong affinity for plutonium and may facilitate its accumulation in the skeleton. Sci Rep 2019; 9:17584. [PMID: 31772265 PMCID: PMC6879641 DOI: 10.1038/s41598-019-53770-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 10/29/2019] [Indexed: 11/09/2022] Open
Abstract
After entering the blood, plutonium accumulates mainly in the liver and the bones. The mechanisms leading to its accumulation in bone are, however, completely unknown. We already know that another uptake pathway not involving the transferrin-mediated pathways is suspected to intervene in the case of the liver. Fetuin, a protein playing an important role in bone metabolism, is proposed as a potential transporter of Pu from serum to bone. For the first time, the binding constants of these two proteins (transferrin and fetuin) with tetravalent plutonium at physiological pH (pH 7.0) were determined by using capillary electrophoresis (CE) coupled with inductively coupled plasma mass spectrometry (ICP-MS). Their very close values (log10 KPuTf = 26.44 ± 0.28 and log10 KPuFet = 26.20 ± 0.24, respectively) suggest that transferrin and fetuin could compete to chelate plutonium, either in the blood or directly at bone surfaces in the case of Pu deposits. We performed competition reaction studies demonstrating that the relative distribution of Pu-protein complexes is fully explained by thermodynamics. Furthermore, considering the average concentrations of transferrin and fetuin in the blood, our calculation is consistent with the bio-distribution of Pu observed in humans.
Collapse
Affiliation(s)
- Claude Vidaud
- CEA, DRF, BIAM-Marcoule, F-30207, Bagnols sur Cèze, France.
| | - Laurent Miccoli
- Laboratoire de RadioToxicologie, CEA, Université de Paris-Saclay, F-91297, Arpajon, France
| | | | | |
Collapse
|
48
|
Bressendorff I, Hansen D, Pasch A, Holt SG, Schou M, Brandi L, Smith ER. The effect of increasing dialysate magnesium on calciprotein particles, inflammation and bone markers: post hoc analysis from a randomized controlled clinical trial. Nephrol Dial Transplant 2019; 36:713-721. [DOI: 10.1093/ndt/gfz234] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Background
The formation of calciprotein particles (CPPs) may be an important component of the humoral defences against ectopic calcification. Although magnesium (Mg) has been shown to delay the transition of amorphous calcium-/phosphate-containing primary CPP (CPP-1) to crystalline apatite-containing secondary CPP (CPP-2) ex vivo, effects on the endogenous CPP pool are unknown.
Methods
We used post hoc analyses from a randomized double-blind parallel-group controlled clinical trial of 28 days treatment with high dialysate Mg of 2.0 mEq/L versus standard dialysate Mg of 1.0 mEq/L in 57 subjects undergoing maintenance hemodialysis for end-stage kidney disease. CPP load, markers of systemic inflammation and bone turnover were measured at baseline and follow-up.
Results
After 28 days of treatment with high dialysate Mg, serum total CPP (−52%), CPP-1 (−42%) and CPP-2 (−68%) were lower in the high Mg group (all P < 0.001) but were unchanged in the standard dialysate Mg group. Tumour necrosis factor-α (−20%) and interleukin-6 (−22%) were also reduced with high dialysate Mg treatment (both P < 0.01). High dialysate Mg resulted in higher levels of bone-specific alkaline phosphatase (a marker of bone formation) (+17%) but lower levels of tartrate-resistant acid phosphatase 5 b (a marker of bone resorption; −33%) (both P < 0.01). Inflammatory cytokines and bone turnover markers were unchanged in the standard dialysate Mg group over the same period.
Conclusions
In this exploratory analysis, increasing dialysate Mg was associated with reduced CPP load and systemic inflammation and divergent changes in markers of bone formation and resorption.
Collapse
Affiliation(s)
- Iain Bressendorff
- Department of Endocrinology and Nephrology, Nordsjællands Hospital, Hillerød, Denmark
- Department of Nephrology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Ditte Hansen
- Department of Nephrology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Andreas Pasch
- Calciscon AG, Nidau, Switzerland
- Insitute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Stephen G Holt
- Department of Nephrology, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Morten Schou
- Department of Cardiology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Lisbet Brandi
- Department of Endocrinology and Nephrology, Nordsjællands Hospital, Hillerød, Denmark
| | - Edward R Smith
- Department of Nephrology, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Calcium Phosphate Bions Cause Intimal Hyperplasia in Intact Aortas of Normolipidemic Rats through Endothelial Injury. Int J Mol Sci 2019; 20:ijms20225728. [PMID: 31731607 PMCID: PMC6888620 DOI: 10.3390/ijms20225728] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Calcium phosphate bions (CPBs) are formed under blood supersaturation with calcium and phosphate owing to the mineral chaperone fetuin-A and representing mineralo-organic particles consisting of bioapatite and multiple serum proteins. While protecting the arteries from a rapid medial calcification, CPBs cause endothelial injury and aggravate intimal hyperplasia in balloon-injured rat aortas. Here, we asked whether CPBs induce intimal hyperplasia in intact rat arteries in the absence of cardiovascular risk factors. Normolipidemic Wistar rats were subjected to regular (once/thrice per week over 5 weeks) tail vein injections of either spherical (CPB-S) or needle-shaped CPBs (CPB-N), magnesium phosphate bions (MPBs), or physiological saline (n = 5 per group). Neointima was revealed in 3/10 and 4/10 rats which received CPB-S or CPB-N, respectively, regardless of the injection regimen or blood flow pattern in the aortic segments. In contrast, none of the rats treated with MPBs or physiological saline had intimal hyperplasia. The animals also did not display signs of liver or spleen injury as well as extraskeletal calcium deposits. Serum alanine/aspartate transaminases, interleukin-1β, MCP-1/CCL2, C-reactive protein, and ceruloplasmin levels did not differ among the groups. Hence, CPBs may provoke intimal hyperplasia via direct endothelial injury regardless of their shape or type of blood flow.
Collapse
|
50
|
|