1
|
Wu H, Shao C, Shi J, Hu Z, Zhou Y, Chen Z, Tang R, Xie Z, Jin W. Hyaluronic acid-mediated collagen intrafibrillar mineralization and enhancement of dentin remineralization. Carbohydr Polym 2023; 319:121174. [PMID: 37567692 DOI: 10.1016/j.carbpol.2023.121174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/01/2023] [Indexed: 08/13/2023]
Abstract
Non-collagenous proteins (NCPs) in the extracellular matrix (ECM) of bone and dentin are known to play a critical regulatory role in the induction of collagen fibril mineralization and are embedded in hyaluronic acid (HA), which acts as a water-retaining glycosaminoglycan and provides necessary biochemical and biomechanical cues. Our previous study demonstrated that HA could regulate the mineralization degree and mechanical properties of collagen fibrils, yet its kinetics dynamic mechanism on mineralization is under debate. Here, we further investigated the role of HA on collagen fibril mineralization and the possible mechanism. The HA modification can significantly promote intrafibrillar collagen mineralization by reducing the electronegativity of the collagen surface to enhance calcium ions (Ca2+) binding capacity to create a local higher supersaturation. In addition, the HA also provides additional nucleation sites and shortens the induction time of amorphous calcium phosphate (ACP)-mediated hydroxyapatite (HAP) crystallization, which benefits mineralization. The acceleration effect of HA on intrafibrillar collagen mineralization is also confirmed in collagen hydrogel and in vitro dentin remineralization. These findings offer a physicochemical view of the regulation effect of carbohydrate polymers in the body on biomineralization, the fine prospect for an ideal biomaterial to repair collagen-mineralized tissues.
Collapse
Affiliation(s)
- Haiyan Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Changyu Shao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Jue Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zihe Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Yanyan Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhuo Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Wenjing Jin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
2
|
Jin C, Zong Y. The role of hyaluronan in renal cell carcinoma. Front Immunol 2023; 14:1127828. [PMID: 36936902 PMCID: PMC10019822 DOI: 10.3389/fimmu.2023.1127828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is associated with high mortality rates worldwide and survival among RCC patients has not improved significantly in the past few years. A better understanding of the pathogenesis of RCC can enable the development of more effective therapeutic strategies against RCC. Hyaluronan (HA) is a glycosaminoglycan located in the extracellular matrix (ECM) that has several roles in biology, medicine, and physiological processes, such as tissue homeostasis and angiogenesis. Dysregulated HA and its receptors play important roles in fundamental cellular and molecular biology processes such as cell signaling, immune modulation, tumor progression and angiogenesis. There is emerging evidence that alterations in the production of HA regulate RCC development, thereby acting as important biomarkers as well as specific therapeutic targets. Therefore, targeting HA or combining it with other therapies are promising therapeutic strategies. In this Review, we summarize the available data on the role of abnormal regulation of HA and speculate on its potential as a therapeutic target against RCC.
Collapse
Affiliation(s)
- Chenchen Jin
- Zhejiang Academy of Science & Technology for Inspection & Quarantine, Hangzhou, Zhejiang, China
| | - Yunfeng Zong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Unravelling the Complex Relationship between Diet and Nephrolithiasis: The Role of Nutrigenomics and Nutrigenetics. Nutrients 2022; 14:nu14234961. [PMID: 36500991 PMCID: PMC9739708 DOI: 10.3390/nu14234961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Nephrolithiasis is an increasingly prevalent condition, especially in high income countries, and is associated with high morbidity. Extraordinary progress in genetics made the identification of genetic forms of nephrolithiasis possible. These genetic diseases are usually rare and do not account for the most common forms of nephrolithiasis that are the result of several factors such as environment, dietary habits, and predisposing genes. This knowledge has shaped what we classify as nephrolithiasis, a condition that is now recognized as systemic. How and to what extent all these factors interact with one another and end in kidney stone formation, growth, and recurrence is not completely understood. Two new research fields have recently been trying to give some answers: nutrigenomics and nutrigenetics. These fields have the aim of understanding the intricate diet/genome interface that influences gene expression regulation mainly through epigenetic mechanisms and results in specific medical conditions such as cancer, metabolic syndrome, and cardiovascular diseases. Epigenetics seems to play a crucial role and could represent the link between environmental factors, that we are constantly exposed to, and risk factors for nephrolithiasis. In this systematic review, we summarize all the available evidence of proven or hypothesized epigenetic mechanisms related to nephrolithiasis.
Collapse
|
4
|
Todorov LG, Sivaguru M, Krambeck AE, Lee MS, Lieske JC, Fouke BW. GeoBioMed perspectives on kidney stone recurrence from the reactive surface area of SWL-derived particles. Sci Rep 2022; 12:18371. [PMID: 36319741 PMCID: PMC9626463 DOI: 10.1038/s41598-022-23331-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/29/2022] [Indexed: 11/18/2022] Open
Abstract
Shock wave lithotripsy (SWL) is an effective and commonly applied clinical treatment for human kidney stones. Yet the success of SWL is counterbalanced by the risk of retained fragments causing recurrent stone formation, which may require retreatment. This study has applied GeoBioMed experimental and analytical approaches to determine the size frequency distribution, fracture patterns, and reactive surface area of SWL-derived particles within the context of their original crystal growth structure (crystalline architecture) as revealed by confocal autofluorescence (CAF) and super-resolution autofluorescence (SRAF) microscopy. Multiple calcium oxalate (CaOx) stones were removed from a Mayo Clinic patient using standard percutaneous nephrolithotomy (PCNL) and shock pulse lithotripsy (SPL). This produced approximately 4-12 mm-diameter PCNL-derived fragments that were experimentally treated ex vivo with SWL to form hundreds of smaller particles. Fractures propagated through the crystalline architecture of PCNL-derived fragments in a variety of geometric orientations to form rectangular, pointed, concentrically spalled, and irregular SWL-derived particles. Size frequency distributions ranged from fine silt (4-8 μm) to very fine pebbles (2-4 mm), according to the Wentworth grain size scale, with a mean size of fine sand (125-250 μm). Importantly, these SWL-derived particles are smaller than the 3-4 mm-diameter detection limit of clinical computed tomography (CT) techniques and can be retained on internal kidney membrane surfaces. This creates clinically undetectable crystallization seed points with extremely high reactive surface areas, which dramatically enhance the multiple events of crystallization and dissolution (diagenetic phase transitions) that may lead to the high rates of CaOx kidney stone recurrence after SWL treatment.
Collapse
Affiliation(s)
- Lauren G. Todorov
- grid.35403.310000 0004 1936 9991Department of Geology, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Mayandi Sivaguru
- grid.35403.310000 0004 1936 9991Cytometry and Microscopy to Omics Facility, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Amy E. Krambeck
- grid.66875.3a0000 0004 0459 167XDepartment of Urology, Mayo Clinic, Rochester, MN USA ,grid.16753.360000 0001 2299 3507Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Matthew S. Lee
- grid.16753.360000 0001 2299 3507Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - John C. Lieske
- grid.66875.3a0000 0004 0459 167XDivision of Nephrology and Hypertension, Mayo Clinic, Rochester, MN USA ,grid.66875.3a0000 0004 0459 167XDepartment of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Bruce W. Fouke
- grid.35403.310000 0004 1936 9991Department of Geology, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Department of Evolution, Ecology and Behavior, University of Illinois at Urbana-Champaign, Urbana, IL USA
| |
Collapse
|
5
|
Sivaguru M, Fouke BW. Renal Macrophages and Multinucleated Giant Cells: Ferrymen of the River Styx? KIDNEY360 2022; 3:1616-1619. [PMID: 36245644 PMCID: PMC9528364 DOI: 10.34067/kid.0003992022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/22/2022] [Indexed: 06/16/2023]
Affiliation(s)
- Mayandi Sivaguru
- Cytometry and Microscopy to Omics Facility, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Bruce W. Fouke
- Cytometry and Microscopy to Omics Facility, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Biocomplexity Theme, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Geology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Evolution, Ecology and Behavior, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
6
|
Yifan Z, Luming S, Wei C, Luwei X, Zheng X, Ruipeng J. Cystine crystal-induced reactive oxygen species associated with NLRP3 inflammasome activation: implications for the pathogenesis of cystine calculi. Int Urol Nephrol 2022; 54:3097-3106. [PMID: 36085346 DOI: 10.1007/s11255-022-03347-6] [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: 05/26/2022] [Accepted: 08/19/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE To investigate whether cystine crystal-induced production of reactive oxygen species (ROS) and activation of NLRP3 inflammasome contribute to cystine calculi formation. METHODS Slc7a9-knockout rats were created as cystine calculi animal models. Kidney histological examination using TEM and immunohistochemistry were performed. The protein expression of NLRP3 and IL-1β and the concentrations of oxidative stress markers such as ROS, MDA and H2O2 in kidney tissues were estimated. In parallel, HK-2 human renal proximal tubule cells were exposed to cystine crystals and NAC treatment. The protein and mRNA expression levels of NLRP3 were evaluated. Finally, cell apoptosis and cystine crystal adherence were also assessed. RESULTS Activation of the NLRP3 inflammasome and marked elevations in MDA, H2O2 and ROS levels were observed both in vivo and in vitro. In particular, the protein and mRNA expression of NLRP3 was significantly increased by cystine crystals, but could be restored by an inhibitor of ROS. In addition, cell apoptosis and cystine crystal adherence were promoted by the NLRP3 inflammasome. The expression of CD44, OPN and HA in HK-2 cells was markedly increased by cystine crystals, but could be decreased by NLRP3 siRNA treatment. CONCLUSION Notably, we found that the activation of NLRP3 by cystine crystal-induced ROS production was of major importance in the pathogenesis of cystine calculi formation.
Collapse
Affiliation(s)
- Zhang Yifan
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Shen Luming
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, 121#, Jiangjiayuan, Nanjing, 210000, China
| | - Chen Wei
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Xu Luwei
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Xu Zheng
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China
| | - Jia Ruipeng
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, China.
| |
Collapse
|
7
|
Vinegar reduced renal calcium oxalate stones by regulating acetate metabolism in gut microbiota and crystal adhesion in rats. Int Urol Nephrol 2022; 54:2485-2495. [DOI: 10.1007/s11255-022-03259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/30/2022] [Indexed: 10/17/2022]
|
8
|
Boadi EA, Shin S, Gombedza F, Bandyopadhyay BC. Differential biomolecular recognition by synthetic vs. biologically-derived components in the stone-forming process using 3D microfluidics. J Mater Chem B 2021; 10:34-46. [PMID: 34779812 PMCID: PMC9045411 DOI: 10.1039/d1tb01213d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcium phosphate (CaP) biomineralization is the hallmark of extra-skeletal tissue calcification and renal calcium stones. Although such a multistep process starts with CaP crystal formation, the mechanism is still poorly understood due to the complexity of the in vivo system and the lack of a suitable approach to simulate a truly in vivo-like environment. Although endogenous proteins and lipids are engaged with CaP crystals in such a biological process of stone formation, most in vitro studies use synthetic materials that can display differential bioreactivity and molecular recognition by the cellular component. Here, we used our in vitro microfluidic (MF) tubular structure, which is the first completely cylindrical platform, with renal tubular cellular microenvironments closest to the functional human kidney tubule, to understand the precise role of biological components in this process. We systematically evaluated the contribution of synthetic and biological components in the stone-forming process in the presence of dynamic microenvironmental cues that originated due to cellular pathophysiology, which are critical for the nucleation, aggregation, and growth of CaP crystals. Our results show that crystal aggregation and growth were enhanced by immunoglobulin G (IgG), which was further inhibited by etidronic acid due to the chelation of extracellular Ca2+. Interestingly, biogenic CaP crystals from mice urine, when applied with cell debris and non-specific protein (bovine serum albumin), exhibited a more discrete crystal growth pattern, compared to exposure to synthetic CaP crystals under similar conditions. Furthermore, proteins found on those calcium crystals from mice urine produced discriminatory effects on crystal-protein attachment. Specifically, such biogenic crystals exhibited enhanced affinity to the proteins inherent to those crystals. More importantly, a physiological comparison of crystal induction in renal tubular cells revealed that biogenic crystals are less effective at producing a sustained rise in cytosolic Ca2+ compared to synthetic crystals, suggesting a milder detrimental effect to downstream signaling. Finally, synthetic crystal-internalized cells induced more oxidative stress, inflammation, and cellular damage compared to the biogenic crystal-internalized cells. Together, these results suggest that the intrinsic nature of biogenically derived components are appropriate to generate the molecular recognition needed for spatiotemporal effects and are critical towards understanding the process of kidney stone formation.
Collapse
Affiliation(s)
- Eugenia Awuah Boadi
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA
| | - Samuel Shin
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA
| | - Farai Gombedza
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA
| | - Bidhan C. Bandyopadhyay
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington DC, 20037, USA,Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington DC, 20064, USA
| |
Collapse
|
9
|
Polat S, Burak Eral H. Elucidating the role of hyaluronic acid in the structure and morphology of calcium oxalate crystals. ADV POWDER TECHNOL 2021. [DOI: 10.1016/j.apt.2021.08.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
10
|
Singh A, Tandon S, Nandi SP, Kaur T, Tandon C. Downregulation of inflammatory mediators by ethanolic extract of Bergenia ligulata (Wall.) in oxalate injured renal epithelial cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114104. [PMID: 33836258 DOI: 10.1016/j.jep.2021.114104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/05/2021] [Accepted: 04/03/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the Indian traditional system of medicine, Bergenia ligulata (Wall.) Engl. has been used for treatment of urolithiasis. Its efficacious nature has led to its incorporation in various commercial herbal formulations such as Cystone and Neeri which are prescribed for kidney related ailments. AIM OF THE STUDY To assess whether ethanolic extract of B. ligulata can mitigate the cascade of inflammatory responses that cause oxidative stress and ultimately cell death in renal epithelial cells exposed to hyperoxaluric conditions. MATERIAL AND METHODS Bioactivity guided fractionation using solvents of varying polarities was employed to evaluate the potential of the extracts of B. ligulata to inhibit the crystallization process. Modulation of crystal morphology was visualized through Scanning electron microscopy (SEM) analysis. Cell death was assessed using flow cytometry based assays. Alteration in the inflammatory mediators was evaluated using real time PCR and immunocytochemistry. Phytochemical characterization of the ethanolic extract was carried out using FTIR, LC-MS and GC-MS. RESULTS Bioactivity guided fractionation for the assessment of antilithiatic activity revealed dose dependent inhibition of nucleation and aggregation process of calcium oxalate crystals in the presence of various extracts, however ethanolic extract showed maximum inhibition and was chosen for further experiments. Studies on renal epithelial NRK-52E cells showed, cytoprotective efficacy of B. ligulata extract against oxalate injury. SEM anaysis further revealed the potential of the extract to modulate the crystal structure and adhesion to renal cell surface. Exposure of the renal cells to the extract led to conversion of the calcium oxalate monohydrate (COM) crystals to the less injurious calcium oxalate dihydrate (COD) form. Expression analysis for oxidative stress and inflammatory biomarkers in NRK-52E cells revealed up-regulation of Mitogen activated protein kinase (MAPK), Osteopontin (OPN) and Nuclear factor- ĸB (NF-ĸB), in response to calcium oxalate insult; which was drastically reduced in the presence of B. ligulata extract. Flow cytometric evaluation pointed to caspase 3 mediated apoptotic cell death in oxalate injured cells, which was attenuated by B. ligulata extract. CONCLUSION Considering the complex multifactorial etiology of urolithiasis, ethanolic extract from B. ligulata can be a promising option for the management of kidney stones, as it has the potential to limit inflammation and the subsequent cell death.
Collapse
Affiliation(s)
- Anubha Singh
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Shoma Paul Nandi
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | | |
Collapse
|
11
|
Khamchun S, Yoodee S, Thongboonkerd V. Dual modulatory effects of diosmin on calcium oxalate kidney stone formation processes: Crystallization, growth, aggregation, crystal-cell adhesion, internalization into renal tubular cells, and invasion through extracellular matrix. Biomed Pharmacother 2021; 141:111903. [PMID: 34328112 DOI: 10.1016/j.biopha.2021.111903] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 01/26/2023] Open
Abstract
Diosmin is a natural flavone glycoside (bioflavonoid) found in fruits and plants with several pharmacological activities. It has been widely used as a dietary supplement or therapeutic agent in various diseases/disorders. Although recommended, evidence of its protective mechanisms against kidney stone disease (nephrolithiasis/urolithiasis), especially calcium oxalate (CaOx) monohydrate (COM) that is the most common type, remained unclear. In this study, we thus systematically evaluated the effects of diosmin (at 2.5-160 nM) on various stages of kidney stone formation processes, including COM crystallization, crystal growth, aggregation, crystal-cell adhesion, internalization into renal tubular cells and invasion through extracellular matrix (ECM). The results showed that diosmin had dose-dependent modulatory effects on all the mentioned COM kidney stone processes. Diosmin significantly increased COM crystal number and mass during crystallization, but reduced crystal size and growth. While diosmin promoted crystal aggregation, it inhibited crystal-cell adhesion and internalization into renal tubular cells. Finally, diosmin promoted crystal invasion through the ECM. Our data provide evidence demonstrating both inhibiting and promoting effects of diosmin on COM kidney stone formation processes. Based on these dual modulatory activities of diosmin, its anti-urolithiasis role is doubtful and cautions should be made for its use in kidney stone disease.
Collapse
Affiliation(s)
- Supaporn Khamchun
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Department of Medical Technology, School of Allied Health Sciences, University of Phayao, Phayao 56000, Thailand; Unit of Excellence in Integrative Molecular Biomedicine, School of Allied Health Sciences, University of Phayao, Phayao 56000, Thailand
| | - Sunisa Yoodee
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
12
|
Sivaguru M, Saw JJ, Wilson EM, Lieske JC, Krambeck AE, Williams JC, Romero MF, Fouke KW, Curtis MW, Kear-Scott JL, Chia N, Fouke BW. Human kidney stones: a natural record of universal biomineralization. Nat Rev Urol 2021; 18:404-432. [PMID: 34031587 DOI: 10.1038/s41585-021-00469-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 02/04/2023]
Abstract
GeoBioMed - a new transdisciplinary approach that integrates the fields of geology, biology and medicine - reveals that kidney stones composed of calcium-rich minerals precipitate from a continuum of repeated events of crystallization, dissolution and recrystallization that result from the same fundamental natural processes that have governed billions of years of biomineralization on Earth. This contextual change in our understanding of renal stone formation opens fundamentally new avenues of human kidney stone investigation that include analyses of crystalline structure and stratigraphy, diagenetic phase transitions, and paragenetic sequences across broad length scales from hundreds of nanometres to centimetres (five Powers of 10). This paradigm shift has also enabled the development of a new kidney stone classification scheme according to thermodynamic energetics and crystalline architecture. Evidence suggests that ≥50% of the total volume of individual stones have undergone repeated in vivo dissolution and recrystallization. Amorphous calcium phosphate and hydroxyapatite spherules coalesce to form planar concentric zoning and sector zones that indicate disequilibrium precipitation. In addition, calcium oxalate dihydrate and calcium oxalate monohydrate crystal aggregates exhibit high-frequency organic-matter-rich and mineral-rich nanolayering that is orders of magnitude higher than layering observed in analogous coral reef, Roman aqueduct, cave, deep subsurface and hot-spring deposits. This higher frequency nanolayering represents the unique microenvironment of the kidney in which potent crystallization promoters and inhibitors are working in opposition. These GeoBioMed insights identify previously unexplored strategies for development and testing of new clinical therapies for the prevention and treatment of kidney stones.
Collapse
Affiliation(s)
- Mayandi Sivaguru
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Carl Zeiss Labs@Location Partner, Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Jessica J Saw
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Elena M Wilson
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - John C Lieske
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Amy E Krambeck
- Department of Urology, Mayo Clinic, Rochester, MN, USA.,Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James C Williams
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael F Romero
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kyle W Fouke
- Jackson School of Geosciences, University of Texas at Austin, Austin, TX, USA
| | - Matthew W Curtis
- Carl Zeiss Microscopy LLC, One North Broadway, White Plains, NY, USA
| | | | - Nicholas Chia
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Bruce W Fouke
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Carl Zeiss Labs@Location Partner, Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Department of Geology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
13
|
Saw JJ, Sivaguru M, Wilson EM, Dong Y, Sanford RA, Fields CJ, Cregger MA, Merkel AC, Bruce WJ, Weber JR, Lieske JC, Krambeck AE, Rivera ME, Large T, Lange D, Bhattacharjee AS, Romero MF, Chia N, Fouke BW. In Vivo Entombment of Bacteria and Fungi during Calcium Oxalate, Brushite, and Struvite Urolithiasis. KIDNEY360 2021; 2:298-311. [PMID: 35373025 PMCID: PMC8740987 DOI: 10.34067/kid.0006942020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/23/2020] [Indexed: 02/04/2023]
Abstract
Background Human kidney stones form via repeated events of mineral precipitation, partial dissolution, and reprecipitation, which are directly analogous to similar processes in other natural and manmade environments, where resident microbiomes strongly influence biomineralization. High-resolution microscopy and high-fidelity metagenomic (microscopy-to-omics) analyses, applicable to all forms of biomineralization, have been applied to assemble definitive evidence of in vivo microbiome entombment during urolithiasis. Methods Stone fragments were collected from a randomly chosen cohort of 20 patients using standard percutaneous nephrolithotomy (PCNL). Fourier transform infrared (FTIR) spectroscopy indicated that 18 of these patients were calcium oxalate (CaOx) stone formers, whereas one patient formed each formed brushite and struvite stones. This apportionment is consistent with global stone mineralogy distributions. Stone fragments from seven of these 20 patients (five CaOx, one brushite, and one struvite) were thin sectioned and analyzed using brightfield (BF), polarization (POL), confocal, super-resolution autofluorescence (SRAF), and Raman techniques. DNA from remaining fragments, grouped according to each of the 20 patients, were analyzed with amplicon sequencing of 16S rRNA gene sequences (V1-V3, V3-V5) and internal transcribed spacer (ITS1, ITS2) regions. Results Bulk-entombed DNA was sequenced from stone fragments in 11 of the 18 patients who formed CaOx stones, and the patients who formed brushite and struvite stones. These analyses confirmed the presence of an entombed low-diversity community of bacteria and fungi, including Actinobacteria, Bacteroidetes, Firmicutes, Proteobacteria, and Aspergillus niger. Bacterial cells approximately 1 μm in diameter were also optically observed to be entombed and well preserved in amorphous hydroxyapatite spherules and fans of needle-like crystals of brushite and struvite. Conclusions These results indicate a microbiome is entombed during in vivo CaOx stone formation. Similar processes are implied for brushite and struvite stones. This evidence lays the groundwork for future in vitro and in vivo experimentation to determine how the microbiome may actively and/or passively influence kidney stone biomineralization.
Collapse
Affiliation(s)
- Jessica J. Saw
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mayandi Sivaguru
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Elena M. Wilson
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Yiran Dong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Robert A. Sanford
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Geology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Chris J. Fields
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Melissa A. Cregger
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Annette C. Merkel
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - William J. Bruce
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Joseph R. Weber
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - John C. Lieske
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Amy E. Krambeck
- Department of Urology, Mayo Clinic, Rochester, Minnesota
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Marcelino E. Rivera
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Timothy Large
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Dirk Lange
- The Stone Centre at Vancouver General Hospital, Department of Urologic Sciences, University of British Columbia, Jack Bell Research Centre, Vancouver, British Columbia, Canada
| | - Ananda S. Bhattacharjee
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Michael F. Romero
- Department of Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Nicholas Chia
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Urology, Mayo Clinic, Rochester, Minnesota
| | - Bruce W. Fouke
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Geology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Evolution, Ecology and Behavior, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
14
|
Mijangos F, Celaya MA, Gainza FJ, Imaz A, Arana E. SEM-EDX linear scanning: a new tool for morpho-compositional analysis of growth bands in urinary stones. J Biol Inorg Chem 2020; 25:705-715. [PMID: 32488434 DOI: 10.1007/s00775-020-01793-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/18/2020] [Indexed: 11/26/2022]
Abstract
The genesis and growth of calculi are imprinted in their structure, so the pathogenesis of lithiasis could potentially be read via proper analytical techniques. In this study, electron microscopy with energy-dispersive X-ray spectroscopy (SEM-EDX) is used to obtain a description of the morphology and compositional structure of a single bladder stone. This technique establishes the chemical and crystalline architecture of the urolith to assess the effect of the chemical environment on its growth. Scanning electron microscopy-backscattered electrons (SEM-BSE) images clearly show that the stone has a multilayered structure. These layers and Liesegang ring-like structures are characterized by one predominant chemical component but also by slighter compositional changes. The mean crystalline components are determined by X-ray diffraction (DRX), infrared spectroscopy (FT-IR), and Raman analysis (RMN). Elemental analysis along a radial trajectory of the calculus by EDX linear scanning (EDX-LS) also reveals the compositional structure of the layers and the spatial distribution of the main chemical components. EDX-LS data processing reveals concentration profiles that clearly show morpho-compositional growth bands, which correspond to precipitation waves and urinary concentration peaks. The width of the growth bands is independent of the radial position, layer, and element analyzed. We conclude that the bands observed are a consequence of slight changes in the biochemical composition of the urine and consequently reflect a short-term biological cycle of the renal system. This non-specific growth rate suggests that stone formation is a kinetically controlled phenomenon in which promoters of crystal cluster aggregation may have played a key role.
Collapse
Affiliation(s)
- Federico Mijangos
- Department of Chemical Engineering, Faculty of Science and Technology, University of the Basque Country UPV/EHU, P.O. Box 644, 48080, Bilbao, Spain.
| | - Miren Arrate Celaya
- Department of Chemical Engineering, Faculty of Science and Technology, University of the Basque Country UPV/EHU, P.O. Box 644, 48080, Bilbao, Spain
| | | | - Ariane Imaz
- BioCruces Sanitary Research Institute, Cruces University Hospital, Barakaldo, Spain
| | - Eunate Arana
- BioCruces Sanitary Research Institute, Cruces University Hospital, Barakaldo, Spain
| |
Collapse
|
15
|
Lobine D, Ahmed S, Aschner M, Khan H, Mirzaei H, Mahomoodally MF. Antiurolithiatic effects of pentacyclic triterpenes: The distance traveled from therapeutic aspects. Drug Dev Res 2020; 81:671-684. [PMID: 32314397 DOI: 10.1002/ddr.21670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/19/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022]
Abstract
Globally, approximately 12% of the population is inflicted by various types of urolithiasis. Standard treatments are available both to avert and treat urolithiasis, but with significant adverse side effects. Pentacyclic triterpenes represent a group of naturally occurring compounds which holds immense potential as therapeutic for treating kidney stone. This review aims to provide an integrative description on how pentacyclic triterpenes can effectively treat calcium oxalate urolithiasis through various mechanisms such as antioxidant, anti-inflammatory, diuretic, and angiotensin-converting enzyme inhibition. Some of the pentacylic triterpenes which shows promising activities include lupeol, oleanolic acid, betulin, and taraxasterol. Moreover, future perspectives in the development of pentacyclic triterpenes in formulations/drugs for urinary stone prevention are highlighted. It is anticipated that compiled information would serve as a scientific baseline to advocate further investigations on the potential of pentacyclic triterpenes in urolithiasis remediation.
Collapse
Affiliation(s)
- Devina Lobine
- Department of Health Sciences, Faculty of Science, University of Mauritius, Réduit, Mauritius
| | - Salman Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohamad F Mahomoodally
- Department of Health Sciences, Faculty of Science, University of Mauritius, Réduit, Mauritius.,Institute of Research and Development, Duy Tan University, Da Nang, 550000, Vietnam
| |
Collapse
|
16
|
Sivaguru M, Lieske JC, Krambeck AE, Fouke BW. GeoBioMed sheds new light on human kidney stone crystallization and dissolution. Nat Rev Urol 2020; 17:1-2. [PMID: 31676884 DOI: 10.1038/s41585-019-0256-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Mayandi Sivaguru
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Carl Zeiss Labs@Location Partner, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - John C Lieske
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Amy E Krambeck
- Department of Urology, Mayo Clinic, Rochester, MN, USA.,Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bruce W Fouke
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Carl Zeiss Labs@Location Partner, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Department of Geology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
17
|
Boadi EA, Deems NJ, Raub CB, Bandyopadhyay BC. Matting Calcium Crystals by Melamine Improves Stabilization and Prevents Dissolution. CRYSTAL GROWTH & DESIGN 2019; 19:6636-6648. [PMID: 31749663 PMCID: PMC6867689 DOI: 10.1021/acs.cgd.9b01036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Melamine induces calcium phosphate (CaP) and calcium oxalate (CaOx) crystal formation; however, the physicochemical mechanism is not clear. Recently, we found that melamine has a discriminatory effect on CaP, CaOx, and CaP + CaOx (Mixed) crystal dissolution. Thus, to delineate the mechanism, we examined crystal interactions through birefringence analysis and found that CaP becomes increasingly birefringent when bound to melamine, while the birefringence of CaOx decreases when it forms CaOx-melamine cocrystals. We also confirmed the feasibility of such melamine-CaP/CaOx co-crystallization at the nanomicromolar range. Interestingly, ammeline, which is a similar triazine, did not accelerate CaP/CaOx/Mixed crystal formation and growth, indicating the specificity of crystal interaction by melamine. Furthermore, melamine stabilizes the CaP/CaOx/Mixed crystals when exposed to a crystal inhibitor (etidronic acid) or dissolution agents (citrate analogues), while it induces crystal growth by increasing crystal retention, suggesting melamine's interference with conventional dissolution remedies. Morphological and elemental analysis of melamine-CaP/CaOx/Mixed co-crystals using scanning electron microscopy further revealed that melamine harbors such crystals by creating a nucleation site. Finally, we confirmed the physiological relevance of melamine exposure using artificial urine to show the induction, stabilization, and retention of mixed crystals in the presence of crystal-inhibitor/dissolution agent and thus established potential causes of recurrence of kidney stones.
Collapse
Affiliation(s)
- Eugenia Awuah Boadi
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, District of Columbia 20422, United States
| | - Nikolaus J. Deems
- The Vitreous State Laboratory, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, United States
| | - Christopher B. Raub
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, United States
| | - Bidhan C. Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, District of Columbia 20422, United States
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, United States
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington, District of Columbia 20037, United States
| |
Collapse
|
18
|
miRNA-34a inhibits cell adhesion by targeting CD44 in human renal epithelial cells: implications for renal stone disease. Urolithiasis 2019; 48:109-116. [PMID: 31506763 DOI: 10.1007/s00240-019-01155-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023]
Abstract
Nephrolithiasis is a very common disease in which cell-crystal adhesion is an essential mechanism for kidney stone formation. This study has explored the anti-adhesion function of the microRNA, miR-34a, by targeting CD44, a cell surface receptor, in human renal epithelial (HK-2) cells. The expression of CD44 was monitored by qPCR and western blot. A luciferase assay validated the target of miR-34a in CD44 3' UTR. Immunofluorescence staining under confocal microscopy was used to detect the cell-crystal adhesion effects in vitro. Pizzolato staining was performed to examine the adhesion role of miR-34a in vivo. In HK-2 cells, miR-34a was down-regulated and CD44 was up-regulated when exposed to calcium oxalate monohydrate crystals. Moreover, miR-34a negatively regulated the expression of CD44. According to the luciferase report assay, miR-34a direct targeted a binding site in the CD44 3'UTR. In vitro experiments, miR-34a overexpression inhibited CD44 expression and cell-crystals adhesion; whereas CD44 overexpression showed reversed results. Furthermore, miR-34a suppressed cell-crystals adhesion and stone formation in vivo. These findings indicate that miR-34a targets CD44 in HK-2 cells and inhibits cell-crystal adhesion both in vitro and in vivo. Based on these results, miR-34a may be a potential therapeutic target for renal stone disease.
Collapse
|
19
|
Manzoor MAP, Agrawal AK, Singh B, Mujeeburahiman M, Rekha PD. Morphological characteristics and microstructure of kidney stones using synchrotron radiation μCT reveal the mechanism of crystal growth and aggregation in mixed stones. PLoS One 2019; 14:e0214003. [PMID: 30901364 PMCID: PMC6430423 DOI: 10.1371/journal.pone.0214003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/05/2019] [Indexed: 11/19/2022] Open
Abstract
Understanding the mechanisms of kidney stone formation, development patterns and associated pathological features are gaining importance due to an increase in the prevalence of the disease and diversity in the presentation of the stone composition. Based on the microstructural characteristics of kidney stones, it may be possible to explain the differences in the pathogenesis of pure and mixed types of stones. In this study, the microstructure and distribution of mineral components of kidney stones of different mineralogy (pure and mixed types) were analyzed. The intact stones removed from patients were investigated using synchrotron radiation X-ray computed microtomography (SR-μCT) and the tomography slice images were reconstructed representing the density and structure distribution at various elevation planes. Infrared (IR) spectroscopes, X-ray diffraction (XRD) and scanning electron microscopy (SEM) were used to confirm the bulk mineral composition in the thin section stones. Observations revealed differences in the micro-morphology of the kidney stones with similar composition in the internal 3-D structure. Calcium oxalate monohydrate stones showed well-organised layering patterns, while uric acid stones showed lower absorption signals with homogenous inner structure. Distinct mineral phases in the mixed types were identified based on the differential absorption rates. The 3-D quantitative analysis of internal porosity and spatial variation between nine different types of stones were compared. The diversity among the microstructure of similar and different types of stones shows that the stone formation is complex and may be governed by both physiological and micro-environmental factors. These factors may predispose a few towards crystal aggregation and stone growth, while, in others the crystals may not establish stable attachment and/or growth.
Collapse
Affiliation(s)
- Muhammed A. P. Manzoor
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
- Department of Urology, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Ashish K. Agrawal
- Technical Physics Division, Bhabha Atomic Research Centre, Indore-Mumbai, India
| | - Balwant Singh
- Technical Physics Division, Bhabha Atomic Research Centre, Indore-Mumbai, India
| | - M. Mujeeburahiman
- Department of Urology, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Punchappady-Devasya Rekha
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
- * E-mail: ,
| |
Collapse
|
20
|
Zhu W, Zhao Z, Chou F, Zuo L, Liu T, Yeh S, Bushinsky D, Zeng G, Chang C. Loss of the androgen receptor suppresses intrarenal calcium oxalate crystals deposition via altering macrophage recruitment/M2 polarization with change of the miR-185-5p/CSF-1 signals. Cell Death Dis 2019; 10:275. [PMID: 30894518 PMCID: PMC6427030 DOI: 10.1038/s41419-019-1358-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022]
Abstract
Crystals can trigger a wide range of kidney injuries that may link to the development of kidney stones. Infiltrating macrophages may influence hyperoxaluria-induced intrarenal calcium oxalate (CaOx) crystals deposition, yet their linkage to sex hormones remains unclear. Here we demonstrated that suppressing the androgen receptor (AR) expression in renal tubular epithelial cells increased the macrophage recruitment/M2 polarization that may result in enhancing the phagocytosis of intrarenal CaOx crystals. Mechanism dissection suggested that AR can suppress macrophage colony-stimulating factor 1 (CSF-1) expression via increasing miRNA-185-5p expression to suppress the M2 macrophage polarization-mediated intrarenal CaOx crystals phagocytosis. The preclinical study using glyoxylate-induced intrarenal CaOx crystals deposition mouse model revealed that renal tubule-specific AR knockout mice have less intrarenal CaOx crystals deposition with more recruited M2 macrophages in the kidney compared with the wild-type mice. Results from the in vivo rat model using hydroxy-L-proline-induced CaOx crystals deposition also demonstrated that targeting the AR with ASC-J9® suppressed the intrarenal CaOx crystals deposition via increasing the renal macrophage recruitment/M2 polarization. Together, results from multiple preclinical studies using multiple in vitro cell lines and in vivo mouse/rat models all demonstrated that targeting the AR with a small molecule ASC-J9® may function via altering macrophage recruitment/M2 polarization to decrease the intrarenal CaOx crystals deposition, a key phenotype seen in many kidney stone disease patients with hyperoxaluria.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Urology and Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, 510230, Guangzhou, China
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Zhijian Zhao
- Department of Urology and Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, 510230, Guangzhou, China
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Fuju Chou
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Li Zuo
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Tongzu Liu
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - David Bushinsky
- Departments of Medicine, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Guohua Zeng
- Department of Urology and Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, 510230, Guangzhou, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA.
- Sex Hormone Research Center, China Medical University/Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
21
|
Zhai H, Wang L, Putnis CV. Inhibition of Spiral Growth and Dissolution at the Brushite (010) Interface by Chondroitin 4-Sulfate. J Phys Chem B 2019; 123:845-851. [PMID: 30615454 DOI: 10.1021/acs.jpcb.8b11531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Modulation of mineralization and demineralization of calcium phosphates (Ca-Ps) with organic macromolecules is a critical process which prevents human kidney stone disease. As a long unbranched polysaccharide of urinary glycosaminoglycans, chondroitin 4-sulfate (Ch4S) has been shown to play an essential role in inhibiting the formation of kidney stones. However, the mechanism of the role of Ch4S remains poorly understood. Here, we used in situ atomic force microscopy to observe the growth and dissolution of spirals on brushite (CaHPO4·2H2O) (010) surfaces. The results show that Ch4S preferentially inhibits the [101]Cc step growth/dissolution by step pinning. This step-specific effect appears to be related to specific binding of Ch4S to Ca sites, as the observed inhibition is not seen in other crystallographic directions where there are fewer Ca terminations. Moreover, Ch4S promotes an increase in the terrace width of [101̅]Cc by the modification of the interfacial energies of the step edge. These in vitro direct observations of Ch4S modulating brushite mineralization and demineralization reveal a dual control of both step kinetics and interfacial energy.
Collapse
Affiliation(s)
- Hang Zhai
- College of Resources and Environment , Huazhong Agricultural University , Wuhan 430070 , China
| | - Lijun Wang
- College of Resources and Environment , Huazhong Agricultural University , Wuhan 430070 , China
| | - Christine V Putnis
- Institut für Mineralogie , University of Münster , 48149 Münster , Germany.,Department of Chemistry , Curtin University , Perth 6845 , Australia
| |
Collapse
|
22
|
Abstract
Calcium kidney stones are common worldwide. Most are idiopathic and composed of calcium oxalate. Calcium phosphate is present in around 80% and may initiate stone formation. Stone production is multifactorial with a polygenic genetic contribution. Phosphaturia is found frequently among stone formers but until recently received scant attention. This review examines possible mechanisms for the phosphaturia and its relevance to stone formation from a wide angle. There is a striking lack of clinical data. Phosphaturia is associated, but not correlated, with hypercalciuria, increased 1,25 dihydroxy-vitamin D [1,25 (OH)2D], and sometimes evidence of disturbances in proximal renal tubular function. Phosphate reabsorption in the proximal renal tubules requires tightly regulated interaction of many proteins. Paracellular flow through intercellular tight junctions is the major route of phosphate absorption from the intestine and can be reduced therapeutically in hyperphosphatemic patients. In monogenic defects stones develop when phosphaturia is associated with hypercalciuria, generally explained by increased 1,25 (OH)2D production in response to hypophosphatemia. Calcification does not occur in disorders with increased FGF23 when phosphaturia occurs in isolation and 1,25 (OH)2D is suppressed. Candidate gene studies have identified mutations in the phosphate transporters, but in few individuals. One genome-wide study identified a polymorphism of the phosphate transporter gene SLC34A4 associated with stones. Others did not find mutations obviously linked to phosphate reabsorption. Future genetic studies should have a wide trawl and should focus initially on groups of patients with clearly defined phenotypes. The global data should be pooled.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.
| |
Collapse
|
23
|
Khamchun S, Thongboonkerd V. Cell cycle shift from G0/G1 to S and G2/M phases is responsible for increased adhesion of calcium oxalate crystals on repairing renal tubular cells at injured site. Cell Death Discov 2018; 4:106. [PMID: 30774989 PMCID: PMC6374384 DOI: 10.1038/s41420-018-0123-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/24/2018] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
Renal tubular cell injury can enhance calcium oxalate monohydrate (COM) crystal adhesion at the injured site and thus may increase the stone risk. Nevertheless, underlying mechanism of such enhancement remained unclear. In the present study, confluent MDCK renal tubular cell monolayers were scratched to allow cells to proliferate and repair the injured site. At 12-h post-scratch, the repairing cells had significant increases in crystal adhesion capacity and cell proliferation as compared to the control. Cell cycle analysis using flow cytometry demonstrated that the repairing cells underwent cell cycle shift from G0/G1 to S and G2/M phases. Cyclosporin A (CsA) and hydroxyurea (HU) at sub-toxic doses caused cell cycle shift mimicking that observed in the repairing cells. Crystal-cell adhesion assay confirmed the increased crystal adhesion capacity of the CsA-treated and HU-treated cells similar to that of the repairing cells. These findings provide evidence indicating that cell cycle shift from G0/G1 to S and G2/M phases is responsible, at least in part, for the increased adhesion of COM crystals on repairing renal tubular cells at the injured site.
Collapse
Affiliation(s)
- Supaporn Khamchun
- 1Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,2Department of Immunology and Immunology Graduate Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- 1Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,3Center for Research in Complex Systems Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
24
|
Cu-bearing stainless steel reduces cytotoxicity and crystals adhesion after ureteral epithelial cells exposing to calcium oxalate monohydrate. Sci Rep 2018; 8:14094. [PMID: 30237503 PMCID: PMC6148291 DOI: 10.1038/s41598-018-32388-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/13/2018] [Indexed: 12/24/2022] Open
Abstract
Calcium oxalate monohydrate (COM), which is the main component of encrustation, may result in cell membrane injury. In addition, cellular damage is suggested to be the primary event attributing to COM crystal binding. To study the interaction between cells and crystals after incubating with a Cu-bearing stainless steel (316L-Cu SS), MTS and flow cytometric analyses were used to assess the cellular responses. The results confirmed that 316L-Cu SS could inhibit cytotoxicity and cellular apoptosis of ureteral epithelial cells (UECs) after COM treatment. Furthermore, molecular expressions of Cu/Zn superoxide dismutase (CuZnSOD), which were evaluated by western blot analysis and real-time quantitative PCR (qPCR), indicated that 316L-Cu SS could inhibit the oxidative stress attributing to up-regulating of CuZnSOD. Moreover, the crystal adhesion cytokine CD44 was examined with western blot and qPCR, and the corresponding hyaluronic (HA) secreted into the medium was measured by enzyme-linked immunosorbent assay (ELISA). All results were confirmed that the expressions of cells cultured with 316L-Cu SS were down-regulated, demonstrating the inhibitory performance of 316L-Cu SS against crystal adhesion.
Collapse
|
25
|
Mittal A, Tandon S, Singla SK, Tandon C. Modulation of lithiatic injury to renal epithelial cells by aqueous extract of Terminalia arjuna. J Herb Med 2018. [DOI: 10.1016/j.hermed.2018.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Vitamin D and calcium kidney stones: a review and a proposal. Int Urol Nephrol 2018; 51:101-111. [DOI: 10.1007/s11255-018-1965-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022]
|
27
|
Tsunezumi J, Sugiura H, Oinam L, Ali A, Thang BQ, Sada A, Yamashiro Y, Kuro-O M, Yanagisawa H. Fibulin-7, a heparin binding matricellular protein, promotes renal tubular calcification in mice. Matrix Biol 2018; 74:5-20. [PMID: 29730503 DOI: 10.1016/j.matbio.2018.04.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/28/2018] [Accepted: 04/28/2018] [Indexed: 12/11/2022]
Abstract
Ectopic calcification occurs during development of chronic kidney disease and has a negative impact on long-term prognosis. The precise molecular mechanism and prevention strategies, however, are not established. Fibulin-7 (Fbln7) is a matricellular protein structurally similar to elastogenic short fibulins, shown to bind dental mesenchymal cells and heparin. Here, we report that Fbln7 is highly expressed in renal tubular epithelium in the adult kidney and mediates renal calcification in mice. In vitro analysis revealed that Fbln7 bound heparin at the N-terminal coiled-coil domain. In Fbln7-expressing CHO-K1 cells, exogenous heparin increased the release of Fbln7 into conditioned media in a dose-dependent manner. This heparin-induced Fbln7 release was abrogated in CHO-745 cells lacking heparan sulfate proteoglycan or in CHO-K1 cells expressing the Fbln7 mutant lacking the N-terminal coiled-coil domain, suggesting that Fbln7 was tethered to pericellular matrix via this domain. Interestingly, Fbln7 knockout (Fbln7-/-) mice were protected from renal tubular calcification induced by high phosphate diet. Mechanistically, Fbln7 bound artificial calcium phosphate particles (aCPP) implicated in calcification and renal inflammation. Binding was decreased significantly in Fbln7-/- primary kidney cells relative to wild-type cells. Further, overexpression of Fbln7 increased binding to aCPP. Addition of heparin reduced binding between aCPP and wild-type cells to levels of Fbln7-/- cells. Taken together, our study suggests that Fbln7 is a local mediator of calcium deposition and that releasing Fbln7 from the cell surface by heparin/heparin derivatives or Fbln7 inhibitory antibodies may provide a novel strategy to prevent ectopic calcification in vivo.
Collapse
Affiliation(s)
- Jun Tsunezumi
- Department of Medicine, Division of Nephrology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hidekazu Sugiura
- Fourth Department of Internal Medicine, Tokyo Women's Medical University, Tokyo 162-8666, Japan; Department of Nephrology, Division of Medicine, Saiseikai Kurihashi Hospital, Saitama 349-1105, Japan
| | - Lalhaba Oinam
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 305-8577, Japan
| | - Aktar Ali
- Center for Mineral Metabolism and Clinical Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bui Quoc Thang
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; Department of Cardiovascular Surgery, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Aiko Sada
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Yoshito Yamashiro
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan.
| |
Collapse
|
28
|
Mittal A, Tandon S, Singla SK, Tandon C. Cytoprotective and anti-apoptotic role of Terminalia arjuna on oxalate injured renal epithelial cells. Cytotechnology 2017; 69:349-358. [PMID: 28181139 DOI: 10.1007/s10616-017-0065-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 01/05/2017] [Indexed: 11/26/2022] Open
Abstract
Urolithiasis is one of the painful multifactorial disorders caused by metabolic abnormalities influencing the composition of body fluids and urine. The bark of Terminalia arjuna (T. arjuna), very well known in Ayurveda for the treatment of cardiovascular diseases, possesses antioxidant and diuretic activity. The present study was undertaken to investigate the antiurolithiatic efficacy of aqueous extract of bark of T. arjuna on oxalate-induced injury to renal tubular epithelial cells. Madin-Darby canine kidney (MDCK) cells were exposed to 2 mM oxalate for 48 h to evaluate the protective effect of T. arjuna aqueous extract on cell viability, CaOx crystal adherence and apoptotic changes caused by oxalate. The results confirmed that oxalate injured MDCK cells were protected by T. arjuna extract. On treatment with a range concentrations, the cell viability increased in a concentration dependent manner. Moreover, the extract prevented the interaction of the calcium oxalate (CaOx) crystals with the cell surface and reduced the number of apoptotic cells. The current data suggests that T. arjuna bark confers a cytoprotective role and based on our results it could be a potential candidate from natural plant sources against urolithiasis.
Collapse
Affiliation(s)
- Amisha Mittal
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, U.P., 201313, India
| | | | - Chanderdeep Tandon
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Sector - 125, Noida, U.P., 201313, India.
| |
Collapse
|
29
|
Rimer JD, Kolbach-Mandel AM, Ward MD, Wesson JA. The role of macromolecules in the formation of kidney stones. Urolithiasis 2016; 45:57-74. [PMID: 27913854 DOI: 10.1007/s00240-016-0948-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/22/2016] [Indexed: 10/20/2022]
Abstract
The formation of crystal aggregates, one of the critical processes in kidney stone pathogenesis, involves interactions between crystals (predominantly calcium oxalate monohydrate, COM) and urinary constituents (e.g., proteins), which serve as an adhesive "glue" between crystals in stones. To develop a better understanding of the protein-crystal interactions that lead to crystal aggregation, we have measured the effect of model proteins on bulk COM crystal properties as well as their adsorption on crystal surfaces using three synthetic polyanions: poly(aspartic acid) (polyD), poly(glutamic acid) (polyE), and poly(acrylic acid) (polyAA). These anionic macromolecules reduced the amount of COM crystal aggregation in bulk solution to an extent similar to that observed for mixture of proteins from normal urine, with little difference between the polymers. In contrast, the polymers exhibited differences in measures of COM crystal growth. Polycations such as poly(arginine) (polyR) and poly(lysine) (polyK) reduced aggregation weakly and exerted negligible effects on crystal growth. All polyions were found to associate with COM crystal surfaces, as evidenced by changes in the zeta potential of COM crystals in electrophoretic mobility measurements. On the other hand, COM aggregation and possibly growth can be promoted by many binary mixtures of polycations and polyanions, which appeared to be mediated by polymer aggregate formation rather than loss of crystal charge stabilization. Similarly, crystal aggregation promotion behavior can be driven by forming aggregates of weakly charged polyanions, like Tamm-Horsfall protein, suggesting that polymer (protein) aggregation may play a critical role in stone formation. Sensitivity of polyanion-COM crystal surface interactions to the chemical composition of polymer side groups were demonstrated by large differences in crystal aggregation behavior between polyD and polyE, which correlated with atomic force microscopy (AFM) measurements of growth inhibition on various COM surfaces and chemical force microscopy (CFM) measurements of unbinding forces between COM crystal surfaces and AFM tips decorated with either carboxylate or amidinium moieties (mimicking polyanion and polyR side chains, respectively). The lack of strong interaction for polyE at the COM (100) surface compared to polyD appeared to be the critical difference. Finally, the simultaneous presence of polyanions and polycations appeared to alter the ability of polycations to mediate unbinding forces in CFM and promote crystal growth. In summary, polyanions strongly associated with COM surfaces and influenced crystallization, while polycations did not, though important differences were observed based on the physicochemical properties of polyanions. Observations suggest that COM aggregation with both polyanion-polycation mixtures and weakly charged polyanions is promoted by polymer aggregate formation, which plays a critical role in bridging crystal surfaces.
Collapse
Affiliation(s)
- Jeffrey D Rimer
- Department of Chemical and Biomolecular Engineering, University of Houston, 4726 Calhoun Rd, Houston, TX, 77204-4004, USA
| | - Ann M Kolbach-Mandel
- Department of Medicine/Nephrology Division, The Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI, 53226, USA
| | - Michael D Ward
- Department of Chemistry and the Molecular Design Institute, New York University, 100 Washington Square East, New York, NY, 10003-6688, USA
| | - Jeffrey A Wesson
- Department of Medicine/Nephrology Division, Department of Veterans Affairs Medical Center, The Medical College of Wisconsin, 5000 West National Avenue, Milwaukee, WI, 53295, USA.
| |
Collapse
|
30
|
Bhadja P, Lunagariya J, Ouyang JM. Seaweed sulphated polysaccharide as an inhibitor of calcium oxalate renal stone formation. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
31
|
Badrinathan S, Shiju MT, Arya R, Rajesh GN, Viswanathan P. Citrus Bioflavonoids Ameliorate Hyperoxaluria Induced Renal Injury and Calcium Oxalate Crystal Deposition in Wistar Rats. Adv Pharm Bull 2015; 5:419-27. [PMID: 26504765 PMCID: PMC4616898 DOI: 10.15171/apb.2015.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 05/12/2015] [Accepted: 05/14/2015] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Citrus is considered as a medically important plant from ancient times and the bioflavonoids of different variety of citrus fruits were well explored for their biological activities. The study aim was to explore the effect of citrus bioflavonoids (CB) to prevent and cure hyperoxaluria induced urolithiasis. METHODS Twenty four Wistar rats were segregated into 4 Groups. Group 1: Control; Group 2: Urolithic (EG-0.75%); Group 3: Preventive study (EG+CB, day 1-50); Group 4: Curative study (EG+CB, day 30-50). Animals received CB orally (20mg/kg body weight) after performing a toxicity study. RESULTS Urinary risk factors and serum renal function parameters were significantly reduced by CB administration in both preventive and curative study (p<0.001). Hematoxylin & Eosin and von Kossa staining demonstrated that renal protection was offered by CB against EG insult. Immunohistochemical analyses revealed over expression and abnormal localization of THP and NF-κB in urolithic rats, while it was effectively regulated by CB supplementation. CONCLUSION CB prevented and significantly controlled lithogenic factors and CaOx deposition in rats. We propose CB as a potential therapy in management of urolithiasis.
Collapse
Affiliation(s)
- Sridharan Badrinathan
- Renal Research Lab, Centre for Bio Medical Research, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Micheal Thomas Shiju
- Renal Research Lab, Centre for Bio Medical Research, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Ramachandran Arya
- Renal Research Lab, Centre for Bio Medical Research, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Ganesh Nachiappa Rajesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Dhanvantrinagar, Puducherry, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Bio Medical Research, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
- Corresponding author: Pragasam Viswanathan,
| |
Collapse
|
32
|
Yiu AJ, Callaghan D, Sultana R, Bandyopadhyay BC. Vascular Calcification and Stone Disease: A New Look towards the Mechanism. J Cardiovasc Dev Dis 2015; 2:141-164. [PMID: 26185749 PMCID: PMC4501032 DOI: 10.3390/jcdd2030141] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Calcium phosphate (CaP) crystals are formed in pathological calcification as well as during stone formation. Although there are several theories as to how these crystals can develop through the combined interactions of biochemical and biophysical factors, the exact mechanism of such mineralization is largely unknown. Based on the published scientific literature, we found that common factors can link the initial stages of stone formation and calcification in anatomically distal tissues and organs. For example, changes to the spatiotemporal conditions of the fluid flow in tubular structures may provide initial condition(s) for CaP crystal generation needed for stone formation. Additionally, recent evidence has provided a meaningful association between the active participation of proteins and transcription factors found in the bone forming (ossification) mechanism that are also involved in the early stages of kidney stone formation and arterial calcification. Our review will focus on three topics of discussion (physiological influences-calcium and phosphate concentration-and similarities to ossification, or bone formation) that may elucidate some commonality in the mechanisms of stone formation and calcification, and pave the way towards opening new avenues for further research.
Collapse
Affiliation(s)
- Allen J. Yiu
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; E-Mails: (A.J.Y.); (D.C.); (R.S.)
| | - Daniel Callaghan
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; E-Mails: (A.J.Y.); (D.C.); (R.S.)
- Department of Pharmacology and Physiology, Georgetown University, 3900 Reservoir Road, NW, Washington, DC 20007, USA
| | - Razia Sultana
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; E-Mails: (A.J.Y.); (D.C.); (R.S.)
| | - Bidhan C. Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; E-Mails: (A.J.Y.); (D.C.); (R.S.)
- Department of Pharmacology and Physiology, Georgetown University, 3900 Reservoir Road, NW, Washington, DC 20007, USA
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Ross Hall 2300 Eye Street, NW, Washington, DC 20037, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-202-745-8622; Fax: +1-202-462-2006
| |
Collapse
|
33
|
Sun XY, Ouyang JM, Zhu WY, Li YB, Gan QZ. Size-dependent toxicity and interactions of calcium oxalate dihydrate crystals on Vero renal epithelial cells. J Mater Chem B 2015; 3:1864-1878. [PMID: 32262259 DOI: 10.1039/c4tb01626b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Urinary crystals in normal and kidney stone patients often have varying sizes; the interaction between renal epithelial cells and COD crystals generated in the tubular fluid could play an initiating role in the pathophysiology of calcium oxalate nephrolithiasis. This study aims to compare the cytotoxicity of micro/nano-calcium oxalate dihydrate (COD) crystals (50 nm, 100 nm, 600 nm, 3 μm, and 10 μm) toward African green monkey renal epithelial (Vero) cells to reveal the mechanism of kidney stone formation at the molecular and cellular levels. METHODS Vero cells were exposed to COD crystals of varying sizes at a concentration of 200 μg mL-1 for 6 h. The effects of COD crystals on Vero cell viability, apoptosis rate, and cellular biochemical parameters [lactate dehydrogenase (LDH), superoxide dismutase (SOD), reactive oxygen species (ROS), hyaluronic acid (HA), osteopontin (OPN), and mitochondrial membrane potential (Δψm)] were determined using biochemical and morphological analyses. RESULTS Vero cell viability and apoptotic rate were closely associated with the size of COD crystals; lower cell viability and higher apoptosis rate were observed in cells exposed to smaller COD crystal size. The expression of SOD, ROS, HA and OPN also changed in a size-dependent manner after exposure to the five different sizes of COD crystals. The area ratio of the (100) face with a high density of Ca2+ ions to the total surface area was also found to influence the severity of cell injury. Cell injury induced by COD crystals was mainly caused by excessive expression of intracellular ROS and reduction of free-radical scavenger SOD. Moreover, binding of large crystals on the cell membrane surface takes more time to cause cell injury than internalized small-sized crystals. The cell death rate was found to be positively correlated with the amount of internalized COD crystals. CONCLUSIONS although the COD toxicity is often disregarded, the size-dependent cytotoxicity of COD crystals toward Vero cells is demonstrated in this study.
Collapse
Affiliation(s)
- Xin-Yuan Sun
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | | | | | | | | |
Collapse
|
34
|
Evenepoel P, Daenen K, Bammens B, Claes K, Meijers B, Naesens M, Sprangers B, Kuypers D, Lerut E. Microscopic nephrocalcinosis in chronic kidney disease patients. Nephrol Dial Transplant 2015; 30:843-8. [DOI: 10.1093/ndt/gfu400] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 11/28/2014] [Indexed: 01/17/2023] Open
|
35
|
Involvement of VKORC1 in the inhibition of calcium oxalate crystal formation in HK-2 cells. ACTA ACUST UNITED AC 2014; 34:376-381. [DOI: 10.1007/s11596-014-1286-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 05/20/2014] [Indexed: 12/21/2022]
|
36
|
Pathophysiology. Urolithiasis 2014. [DOI: 10.1007/978-1-4614-8196-6_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Aggarwal KP, Narula S, Kakkar M, Tandon C. Nephrolithiasis: molecular mechanism of renal stone formation and the critical role played by modulators. BIOMED RESEARCH INTERNATIONAL 2013; 2013:292953. [PMID: 24151593 PMCID: PMC3787572 DOI: 10.1155/2013/292953] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/26/2013] [Indexed: 12/14/2022]
Abstract
Urinary stone disease is an ailment that has afflicted human kind for many centuries. Nephrolithiasis is a significant clinical problem in everyday practice with a subsequent burden for the health system. Nephrolithiasis remains a chronic disease and our fundamental understanding of the pathogenesis of stones as well as their prevention and cure still remains rudimentary. Regardless of the fact that supersaturation of stone-forming salts in urine is essential, abundance of these salts by itself will not always result in stone formation. The pathogenesis of calcium oxalate stone formation is a multistep process and essentially includes nucleation, crystal growth, crystal aggregation, and crystal retention. Various substances in the body have an effect on one or more of the above stone-forming processes, thereby influencing a person's ability to promote or prevent stone formation. Promoters facilitate the stone formation while inhibitors prevent it. Besides low urine volume and low urine pH, high calcium, sodium, oxalate and urate are also known to promote calcium oxalate stone formation. Many inorganic (citrate, magnesium) and organic substances (nephrocalcin, urinary prothrombin fragment-1, osteopontin) are known to inhibit stone formation. This review presents a comprehensive account of the mechanism of renal stone formation and the role of inhibitors/promoters in calcium oxalate crystallisation.
Collapse
Affiliation(s)
- Kanu Priya Aggarwal
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh 173234, India
| | - Shifa Narula
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh 173234, India
| | - Monica Kakkar
- Department of Biochemistry, Himalyan Institute Hospital Trust, Swami Ram Nagar, Dehradun, Uttrakhand 248140, India
| | - Chanderdeep Tandon
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh 173234, India
| |
Collapse
|
38
|
Djojodimedjo T, Soebadi DM, Soetjipto. Escherichia coli infection induces mucosal damage and expression of proteins promoting urinary stone formation. Urolithiasis 2013; 41:295-301. [PMID: 23756638 DOI: 10.1007/s00240-013-0577-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 05/25/2013] [Indexed: 11/28/2022]
Abstract
The effect of urinary tract infection (UTI) on mucosal damage and production of proteins promoting urinary stone formation has not been elucidated. Osteopontin production, with associated mucosal damage due to UTI, may allow easier crystal retention and nucleation resulting in stone formation. The aim of this study is to demonstrate that expression of osteopontin (OPN), OPN mRNA, TLR-4, JNK, TNFR-1, iNOS, HMGB-1, and apoptosis process is higher than normal at renal tubular cells due to urinary tract infection by Escherichia coli. Adult male New Zealand strain rabbits were used. Thirty New Zealand strain rabbits were divided into three groups. The first group acted as controls, the second group underwent ligation of right ureter, and the third group underwent ligation of right ureter and injection of Escherichia coli 105/ml proximal to ligation. Nephrectomy and histological examination were performed after 5 days. All groups were HE stained to examine mucosal damage, specific monoclonal antibodies for TLR-4, JNK, mRNA OPN, OPN, TNFR-1, iNOS and HMGB-1. Apoptotic nuclei were demonstrated using TUNEL method. Statistical calculations were performed using ANOVA test, with p < 0.05 considered significant. The findings confirmed the hypothesis that infection of urinary tract by Escherichia coli demonstrated higher expression of OPN, OPN mRNA, TLR-4, JNK, TNFR-1, iNOS, HMGB-1, apoptosis process and mucosal damage than normal. Infection of urinary tract by Escherichia coli caused higher than normal expression of promoter protein osteopontin and mucosal damage at renal tubular cells. These suggest that urinary infection may promote stone formation by mucosal damage and elevate promoter protein osteopontin at tubulus cell, allowing easier crystal retention and nucleation.
Collapse
Affiliation(s)
- T Djojodimedjo
- Department of Urology, Airlangga University School of Medicine, Dr. Soetomo Hospital, Jln Moestopo No. 6-8, Surabaya, 60286, East Java, Indonesia.
| | | | | |
Collapse
|
39
|
Oxygen nano-bubble water reduces calcium oxalate deposits and tubular cell injury in ethylene glycol-treated rat kidney. Urolithiasis 2013; 41:279-94. [PMID: 23754513 DOI: 10.1007/s00240-013-0576-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 05/25/2013] [Indexed: 01/05/2023]
Abstract
Renal tubular cell injury induced by oxalate plays an important role in kidney stone formation. Water containing oxygen nano-bubbles (nanometer-sized bubbles generated from oxygen micro-bubbles; ONB) has anti-inflammatory effects. Therefore, we investigated the inhibitory effects of ONB water on kidney stone formation in ethylene glycol (EG)-treated rats. We divided 60 rats, aged 4 weeks, into 5 groups: control, the water-fed group; 100 % ONB, the 100 % ONB water-fed group; EG, the EG treated water-fed group; EG + 50 % ONB and EG + 100 % ONB, water containing EG and 50 % or 100 % ONB, respectively. Renal calcium oxalate (CaOx) deposition, urinary excretion of N-acetyl-β-D-glucosaminidase (NAG), and renal expression of inflammation-related proteins, oxidative stress biomarkers, and the crystal-binding molecule hyaluronic acid were compared among the 5 groups. In the control and 100 % ONB groups, no renal CaOx deposits were detected. In the EG + 50 % ONB and EG + 100 % ONB groups, ONB water significantly decreased renal CaOx deposits, urinary NAG excretion, and renal monocyte chemoattractant protein-1, osteopontin, and hyaluronic acid expression and increased renal superoxide dismutase-1 expression compared with the EG group. ONB water substantially affected kidney stone formation in the rat kidney by reducing renal tubular cell injury. ONB water is a potential prophylactic agent for kidney stones.
Collapse
|
40
|
Abstract
There are differences in renal anatomy and physiology between rabbits and other domestic species. Neurogenic renal ischemia occurs readily. Reversible prerenal azotemia may be seen in conjunction with gut stasis. Potentially fatal acute renal failure may be due to structural kidney damage or post-renal disease. Chronic renal failure is often associated with encephalitozoonosis. Affected rabbits cannot vomit and often eat well. Weight loss, lethargy, and cachexia are common clinical signs. Polydypsia/polyuria may be present. Derangements in calcium and phosphorus metabolism are features of renal disease. Radiography is always indicated. Urolithiasis, osteosclerosis, aortic and renal calcification are easily seen on radiographs.
Collapse
|
41
|
Presores JB, Swift JA. Adhesion properties of uric acid crystal surfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:7401-7406. [PMID: 22530791 DOI: 10.1021/la3010272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Two key steps in kidney stone formation--crystal aggregation and attachment to renal tissues--depend on the surface adhesion properties of the crystalline components. Anhydrous uric acid (UA) is the most common organic crystalline phase found in human kidney stones. Using chemical force microscopy, the adhesion force between various functional groups and the largest (100) surface of UA single crystals was measured in both aqueous solution and model urine. Adhesion trends in the two solutions were identical, but were consistently lower in the latter. Changes in the solution ionic strength and pH were also found to affect the magnitude of the adhesion. UA surfaces showed the strongest adhesion to cationic functionalities, which is consistent with ionization of some surface uric acid molecules to urate. Although hydrogen-bonding and van der Waals interactions are usually considered to be dominant forces in the association between neutral organic compounds, this work demonstrates that electrostatic interactions can be important, particularly when dealing with weak acids under certain solution conditions.
Collapse
Affiliation(s)
- Janeth B Presores
- Department of Chemistry, Georgetown University, 37th and O Streets Northwest, Washington, D.C. 20057-1227, USA
| | | |
Collapse
|
42
|
Stridh S, Palm F, Hansell P. Renal interstitial hyaluronan: functional aspects during normal and pathological conditions. Am J Physiol Regul Integr Comp Physiol 2012; 302:R1235-49. [PMID: 22513743 DOI: 10.1152/ajpregu.00332.2011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The glycosaminoglycan (GAG) hyaluronan (HA) is recognized as an important structural component of the extracellular matrix, but it also interacts with cells during embryonic development, wound healing, inflammation, and cancer; i.e., important features in normal and pathological conditions. The specific physicochemical properties of HA enable a unique hydration capacity, and in the last decade it was revealed that in the interstitium of the renal medulla, where the HA content is very high, it changes rapidly depending on the body hydration status while the HA content of the cortex remains unchanged at very low amounts. The kidney, which regulates fluid balance, uses HA dynamically for the regulation of whole body fluid homeostasis. Renomedullary HA elevation occurs in response to hydration and during dehydration the opposite occurs. The HA-induced alterations in the physicochemical characteristics of the interstitial space affects fluid flux; i.e., reabsorption. Antidiuretic hormone, nitric oxide, angiotensin II, and prostaglandins are classical hormones/compounds involved in renal fluid handling and are important regulators of HA turnover during variations in hydration status. One major producer of HA in the kidney is the renomedullary interstitial cell, which displays receptors and/or synthesis enzymes for the hormones mentioned above. During several kidney disease states, such as ischemia-reperfusion injury, tubulointerstitial inflammation, renal transplant rejection, diabetes, and kidney stone formation, HA is upregulated, which contributes to an abnormal phenotype. In these situations, cytokines and other growth factors are important stimulators. The immunosuppressant agent cyclosporine A is nephrotoxic and induces HA accumulation, which could be involved in graft rejection and edema formation. The use of hyaluronidase to reduce pathologically overexpressed levels of tissue HA is a potential therapeutic tool since diuretics are less efficient in removing water bound to HA in the interstitium. Although the majority of data describing the role of HA originate from animal and cell studies, the available data from humans demonstrate that an upregulation of HA also occurs in diabetic kidneys, in transplant-rejected kidneys, and during acute tubular necrosis. This review summarizes the current knowledge regarding interstitial HA in the role of regulating kidney function during normal and pathological conditions. It encompasses mechanistic insights into the background of the heterogeneous intrarenal distribution of HA; i.e., late nephrogenesis, its regulation during variations in hydration status, and its involvement during several pathological conditions. Changes in hyaluronan synthases, hyaluronidases, and binding receptor expression are discussed in parallel.
Collapse
Affiliation(s)
- Sara Stridh
- Dept. of Medical Cell Biology, Uppsala Univ., Biomedical Center, PO Box 571, SE-751 23 Uppsala, Sweden
| | | | | |
Collapse
|
43
|
Poon NW, Gohel MDI. Urinary glycosaminoglycans and glycoproteins in a calcium oxalate crystallization system. Carbohydr Res 2011; 347:64-8. [PMID: 22119438 DOI: 10.1016/j.carres.2011.09.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/20/2011] [Accepted: 09/21/2011] [Indexed: 11/29/2022]
Abstract
This study measures the effects of total urinary glycosaminoglycans (GAGs), glycoproteins (GPs) and individual GAGs on the nucleation rates (Bo), growth rates (G) and suspension densities (Mт) of calcium oxalate (CaOx) crystallization by the mixed suspension mixed product removal (MSMPR) system. Total urinary GAGs, glycoproteins and individual GAGs including heparan sulfate (HS), chondroitin sulfate (CS) and Hyaluronic acid (HA) were added into the artificial urine (AU) and then introduced into the MSMPR test chamber and the crystal sizes and numbers were analyzed by a particle counter. The effects of added GAGs and GPs on CaOx crystallization were reflected by the changes on the crystallization indexes including the Bo, G and Mт of CaOx that were calculated based on the crystal size and numbers. Total urinary GAGs showed no statistical significance on CaOx crystallization. However, individual GAGs such as HA, CS and HS enhanced Bo and suppressed the G when measured individually. CS and HS enhanced the Mт while HA shown no significant change in the Mт of CaOx. Total urinary GPs showed an increase in the G and Mт of crystals. Although total urinary GAGs showed no statistically significant effect on CaOx crystallization, individual GAGs (CS, HS) promoted the CaOx crystallization by increasing the suspension density of smaller crystals, indicative of reduced risk of stones while HA showed no significance in the M(T) of CaOx formed. Urinary GPs indicated increased sizes and M(T) suggesting larger crystals and/or aggregates.
Collapse
Affiliation(s)
- Ngork Wah Poon
- Dept. of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | | |
Collapse
|
44
|
Thurgood LA, Sørensen ES, Ryall RL. The effect of intracrystalline and surface-bound osteopontin on the attachment of calcium oxalate dihydrate crystals to Madin-Darby canine kidney (MDCK) cells in ultrafiltered human urine. BJU Int 2011; 109:1100-9. [DOI: 10.1111/j.1464-410x.2011.10530.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Renal epithelial cell injury and its promoting role in formation of calcium oxalate monohydrate. J Biol Inorg Chem 2010; 16:405-16. [PMID: 21127923 DOI: 10.1007/s00775-010-0738-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 10/27/2010] [Indexed: 10/18/2022]
Abstract
The injurious effect of hydrogen peroxide (H(2)O(2)) on renal epithelial cells of the African green monkey (Vero cells) and the difference in the modulation of Vero cells on crystal growth of calcium oxalate (CaOxa) before and after injury were investigated. The degree of injury of Vero cells was proportional to the concentration and action time of H(2)O(2). After the cells had been injured, the released amount of malonaldehyde in the culture medium increased, the superoxide dismutase activity decreased, the expression quantity of osteopontin on the surface of Vero cells increased significantly, the zeta potential became more negative, and the amount of CaOxa crystals adhering to cells increased. The CaOxa crystals induced by the cells in the control group were round and blunt; however, those induced by the injured cells had irregular shapes with sharp edges and corners. As the crystallization time increased from 6 to 24 h, the size of the crystals induced by the injured cells increased accordingly, whereas that of crystals induced by the control cells did not increase significantly. The injured cells could promote the growth of CaOxa crystals and their adhesion to the cells; thus, the formation of CaOxa stones was promoted. The cells in the control group could also be injured after being incubated with supersaturated CaOxa solution for a long time, which promoted the crystallization of CaOxa. The results suggest that the retention of supersaturated CaOxa solution or CaOxa crystals in the urinary tract for a long time is a risk factor for the formation of kidney stones.
Collapse
|
46
|
Yuen JWM, Gohel MDI, Poon NW, Shum DKY, Tam PC, Au DWT. The initial and subsequent inflammatory events during calcium oxalate lithiasis. Clin Chim Acta 2010; 411:1018-26. [PMID: 20347754 DOI: 10.1016/j.cca.2010.03.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 03/11/2010] [Accepted: 03/12/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Crystallization is believed to be the initiation step of urolithiasis, even though it is unknown where inside the nephron the first crystal nucleation occurs. METHODS Direct nucleation of calcium oxalate and subsequent events including crystal retention, cellular damage, endocytosis, and hyaluronan (HA) expression, were tested in a two-compartment culture system with intact human proximal tubular HK-2 cell monolayer. RESULTS Calcium oxalate dihydrate (COD) was nucleated and bound onto the apical surface of the HK-2 cells under hypercalciuric and hyperoxaluric conditions. These cells displayed mild cellular damage and internalized some of the adhered crystals within 18h post-COD-exposure, as revealed by electron microscopy. Prolonged incubation in complete medium caused significant damage to disrupt the monolayer integrity. Furthermore, hyaluronan disaccharides were detected in the harvested media, and were associated with HAS-3 mRNA expression. CONCLUSION Human proximal cells were able to internalize COD crystals which nucleated directly onto the apical surface, subsequently triggering cellular damage and HAS-3 specific hyaluronan synthesis as an inflammatory response. The proximal tubule cells here demonstrate that it plays an important role in facilitating urolithiasis via endocytosis and creating an inflammatory environment whereby free hyaluronan in tubular fluid can act as crystal-binding molecule at the later segments of distal and collecting tubules.
Collapse
Affiliation(s)
- John W M Yuen
- Department of Health Technology & Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | | | | | | | | | | |
Collapse
|
47
|
MacDougall L, Taheri S, Crofton P. Biochemical risk factors for stone formation in a Scottish paediatric hospital population. Ann Clin Biochem 2010; 47:125-30. [DOI: 10.1258/acb.2009.009146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Renal stones in children, although rare, may be associated with morbidity and renal damage. Scottish children have a different ethnic composition and diet compared with paediatric populations previously studied. Urinary stone promoters include calcium, oxalate and urate. Postulated inhibitors include citrate and glycosaminoglycans (GAGs). We tested the hypothesis that Scottish paediatric stone-formers have higher excretion of urinary stone promoters (calcium/oxalate/urate) and/or lower excretion of stone inhibitors (citrate/GAGs) than children with isolated haematuria and controls. Methods In this case-controlled study, we measured creatinine, calcium, oxalate, urate, citrate and GAGs in random urine samples from 24 stone-formers (excluding inherited metabolic disorders), median age 10.2 (range 1.0–17.2) y; 25 patients with isolated haematuria, 6.3 (0.6–13.7) y; and 32 controls, 7.5 (0.8–14.7) y. Results Excretion of urinary promoters and inhibitors differed among stone-formers, haematuria and control groups for (median (range)): calcium (0.82 (0.02–2.19), 0.43 (0.08–2.65), 0.31 (0.04–2.12) mmol/mmol creatinine, respectively, P = 0.005), citrate (0.42 (0.13–0.72), 0.33 (0.05–0.84), 0.61 (0.11–1.75) mmol/mmol creatinine, P = 0.001), calcium:citrate ratio (1.68 (0.19–4.81), 1.30 (0.19–9.57), 0.54 (0.10–2.27) mmol/mmol, P < 0.0001) and the promoter:inhibitor ratio (calcium × oxalate)/(citrate × GAGs) (8.3 (1.0–82.5), 4.3 (1.2–69.5), 2.8 (0.3–13.2) mmol/g, P < 0.0001). Conclusions Scottish paediatric stone-formers had lower urinary citrate excretion and higher urinary calcium excretion, calcium:citrate ratio and promoter:inhibitor ratio compared with controls. Urinary calcium excretion and promoter:inhibitor ratio was also higher than children with isolated haematuria. Nevertheless, marked overlap between the stone-former and haematuria groups for promoter:inhibitor and calcium:citrate ratios suggests that some patients with isolated haematuria may be at future risk of urolithiasis.
Collapse
Affiliation(s)
| | - Sepideh Taheri
- Department of Medical Paediatrics, Royal Hospital for Sick Children, Edinburgh, Scotland, UK
| | | |
Collapse
|
48
|
Evenepoel P, Lerut E, Naesens M, Bammens B, Claes K, Kuypers D, Vermeersch P, Meijers B, Van Damme B, Vanrenterghem Y. Localization, etiology and impact of calcium phosphate deposits in renal allografts. Am J Transplant 2009; 9:2470-8. [PMID: 19681815 DOI: 10.1111/j.1600-6143.2009.02792.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hypercalcemia, hypophosphatemia and renal phosphate wasting are common after kidney transplantation. Animal data suggest that these alterations in mineral metabolism may contribute to calcium phosphate (CaPhos) deposition in the kidney and renal dysfunction. We tested the hypothesis that CaPhos deposition is highly prevalent in the early posttransplant period and is related to a disturbed mineral metabolism. For this purpose, biomarkers of mineral metabolism and renal calcium and phosphorus handling were prospectively assessed in 201 renal transplant recipients. CaPhos deposits were observed in 4.6, 30.4 and 24.7% of protocol biopsies obtained at the time of engraftment, and 3 and 12 months thereafter, respectively. In multivariate logistic regression analysis, high calcium and low serum phosphorus levels were independently associated with renal CaPhos deposition at month 3. The extent of CaPhos deposition correlated significantly with the severity of mineral metabolism disturbances. Renal function after a mean follow-up of 33 months was similar in patients with and without CaPhos deposition at month 3. In conclusion, our data demonstrate that CaPhos deposition is highly prevalent in the early posttransplant period and suggest that a disordered mineral metabolism is implicated in its pathogenesis. The clinical relevance of CaPhos deposition remains to be established.
Collapse
Affiliation(s)
- P Evenepoel
- Department of Medicine, Division of Nephrology, University Hospital Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Grover PK, Thurgood LA, Wang T, Ryall RL. The effects of intracrystalline and surface-bound proteins on the attachment of calcium oxalate monohydrate crystals to renal cells in undiluted human urine. BJU Int 2009; 105:708-15. [PMID: 19694711 DOI: 10.1111/j.1464-410x.2009.08816.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To compare the binding to Madin-Darby canine kidney (MDCK)-II cells of: (i) inorganic calcium oxalate monohydrate (iCOM) crystals and COM crystals precipitated from urine containing different concentrations of protein; and (ii) urinary COM crystals containing intracrystalline and intracrystalline + surface-bound protein. MATERIALS AND METHODS Urinary COM crystals were generated in sieved (sCOM), centrifuged and filtered (cfCOM), and ultrafiltered (ufCOM) portions of a pooled human urine and their adhesion to MDCK-II cells was compared using six different ultrafiltered urine samples as the binding medium. Crystal matrix extract (CME) was prepared by demineralizing calcium oxalate crystals precipitated from human urine and used to prepare COM crystals with intracrystalline, and intracrystalline + surface-bound CME at protein concentrations of 0, 0.05, 0.1, 0.5 and 5.0 mg/L. The amount of protein associated with the crystals was qualitatively assessed by sodium dodecyl sulphate-polyacrylamide gel electrophoresis and Western blotting, using prothrombin fragment 1 (PTF1) as a marker. Protein concentration was determined in sieved, centrifuged and filtered, and ultrafiltered fractions of 10 additional urine samples. RESULTS The median crystal attachment in the six urine types decreased in the order iCOM > ufCOM > cfCOM = sCOM, in inverse proportion to the concentration of protein in the solution or urine from which they were precipitated. sCOM and cfCOM crystals bound approximately 23% less than iCOM crystals. The attachment of COM crystals generated in the presence of increasing concentrations of CME proteins was unaffected up to a concentration of 5 mg/L, but binding of crystals containing the same concentrations of intracrystalline + surface-bound proteins decreased proportionally at protein concentrations from 0 to 5.0 mg/L. CONCLUSION Inorganic COM crystals bind significantly more strongly to MDCK-II cells than urinary crystals precipitated from sieved, centrifuged and filtered, and ultrafiltered urine, and binding affinity is inversely related to the concentration of protein in the urine in which they are formed. While both intracrystalline and superficial CME proteins reduce the attachment of COM crystals to MDCK-II cells, those located on the crystal surface have a greater influence than those incarcerated within the mineral bulk. Future cell-crystal interaction studies should use urinary crystals and be performed in human urine.
Collapse
Affiliation(s)
- Phulwinder K Grover
- Urology Unit, Department of Surgery, School of Medicine, The Flinders University of South Australia, Bedford Park, South Australia, Australia.
| | | | | | | |
Collapse
|
50
|
Heiene R, Rumsby G, Ziener M, Dahl SA, Tims C, Teige J, Ottesen N. Chronic kidney disease with three cases of oxalate-like nephrosis in Ragdoll cats. J Feline Med Surg 2009; 11:474-80. [PMID: 19095478 PMCID: PMC10832842 DOI: 10.1016/j.jfms.2008.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2008] [Indexed: 10/21/2022]
Abstract
Two unrelated Ragdoll cat mothers in Norway were found dead from renal disease. The histopathology was consistent with oxalate nephrosis with chronic or acute-on-chronic underlying kidney disease. Both cats had offspring and relatives with signs of urinary tract disease, including a kitten dead with urethral gravel. Eleven living Ragdoll cats, including nine relatives of the dead cats and the male father of a litter with similarly affected animals, were tested for primary hyperoxaluria (PH) type 1 and 2 by urine oxalate and liver enzyme analysis. Renal ultrasound revealed abnormalities in five living cats. One of these was azotaemic at the time of examination and developed terminal kidney disease 9 months later. A diagnosis of PH was excluded in 11 cats tested. The inheritance and aetiological background of the renal disease present in the breed remains unresolved at this point in time.
Collapse
MESH Headings
- Animals
- Cat Diseases/diagnostic imaging
- Cat Diseases/genetics
- Cat Diseases/pathology
- Cat Diseases/urine
- Cats
- Female
- Hyperoxaluria, Primary/complications
- Hyperoxaluria, Primary/diagnostic imaging
- Hyperoxaluria, Primary/pathology
- Hyperoxaluria, Primary/urine
- Hyperoxaluria, Primary/veterinary
- Kidney Failure, Chronic/diagnostic imaging
- Kidney Failure, Chronic/etiology
- Kidney Failure, Chronic/pathology
- Kidney Failure, Chronic/urine
- Kidney Failure, Chronic/veterinary
- Male
- Nephrosis/complications
- Nephrosis/diagnostic imaging
- Nephrosis/pathology
- Nephrosis/urine
- Nephrosis/veterinary
- Norway
- Oxalates/urine
- Ultrasonography
Collapse
Affiliation(s)
- Reidun Heiene
- Department of Companion Animal Clinical Sciences, Norwegian School of Veterinary Science, N-0033 Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|