1
|
Wu X, Liu X, Song Y. The association of glyphosate exposure with kidney stones in American adults: A nationally representative cross-sectional study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117189. [PMID: 39405966 DOI: 10.1016/j.ecoenv.2024.117189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE Glyphosate has been ubiquitously present in our living environment due to its efficient herbicidal ability, but its association with the prevalence of kidney stones remains uncertain. This study aims to explore the impact of glyphosate exposure on kidney stones and to investigate the mediating effects of some serologic indicators. Furthermore, we attempt to identify the specific populations at greater risk of exposure. METHODS This is a cross-sectional study of the U.S. adult population examining the association between glyphosate exposure and kidney stones based on data from the 2013-2018 National Health and Nutrition Examination Survey (NHANES). We implemented multi-model-adjusted logistic regression and smoothed curve fitting to explore the connection between them. Further subgroup analyses were conducted to confirm the magnitude of exposure risk in specific populations. Mediation effects analysis served to provide insight into the underlying mechanisms of the link. RESULTS A total of 4302 participants' health data were ultimately analyzed, and the prevalence of kidney stones was 10.85 %. Participants with the highest urinary glyphosate(uGLY) content(Q3) had a higher prevalence of kidney stones compared with participants with the lowest uGLY content(Q1) (OR=1.70, 95 %CI: 1.10-2.63). Smoothed curve fitting revealed a linear positive association between ln-transformed uGLY and kidney stones (OR=1.21,95 %CI:1.08-1.37, LLR=0.291), and this exposure-outcome effect was at greater risk in men (OR=1.24,95 %CI: 1.05-1.46), non-Hispanic whites (OR=1.29, 95 %CI: 1.09-1.53), and hypertensive groups (OR=1.23,95 %CI: 1.05-1.44). Serum biochemical markers HDL, ALP, and serum glucose partially mediated the correlation between glyphosate and kidney stones (2.44-4.20 %). CONCLUSION Glyphosate exposure is significantly associated with the prevalence of kidney stones. In men, non-Hispanic whites, and hypertensive populations, the management of glyphosate exposure should be emphasized, and appropriate protective strategies may be beneficial in reducing the burden of kidney stones. More high-quality clinical inquiries and animal toxicology experiments are still required to verify the reliability of our findings and their underlying mechanisms.
Collapse
Affiliation(s)
- Xiaodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xi Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, China
| | - Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
2
|
Sholan R, Aliyev R, Hashimova U, Karimov S, Bayramov E. Urinary Stone Composition Analysis of 1465 Patients: The First Series from Azerbaijan. ARCHIVES OF IRANIAN MEDICINE 2024; 27:618-623. [PMID: 39534996 PMCID: PMC11558612 DOI: 10.34172/aim.32026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Urinary stone disease is a prevalent health issue worldwide, with varying incidence influenced by multiple factors. This study aims to provide the first comprehensive analysis of urinary stone composition in Azerbaijan. METHODS A retrospective study was conducted on 1465 patients, aged 1‒83 years, who underwent biochemical urinary stone analysis at the Department of Renal Diseases and Organ Transplantation, Azerbaijan State Security Service Military Hospital, between April 2015 and December 2023. Stone samples were analyzed using Fourier transform infrared (FTIR) spectroscopy. Statistical analyses were performed using the IBM® SPSS software version 29.0. RESULTS The cohort had a median age of 45 years, with a male-to-female ratio of 1.65:1. Calcium oxalate stones were the most common (56.2%), followed by uric acid (33.7%), struvite (5.3%), cystine (2.5%), calcium phosphate (1.9%), and xanthine (0.1%) stones. Men had a higher prevalence of calcium oxalate and uric acid stones, while women had more struvite stones. Mixed stones were common, particularly among uric acid and struvite stones. Significant differences in stone composition were observed between age groups and genders, with uric acid stones found predominantly in older individuals. CONCLUSION This study highlights the predominance of calcium oxalate stones and the elevated prevalence of uric acid stones in Azerbaijan, emphasizing the need for tailored diagnostic and therapeutic approaches. The high prevalence of mixed stones underscores the complexity of urinary stone disease and the need for comprehensive metabolic evaluation and individualized preventive strategies.
Collapse
Affiliation(s)
- Rashad Sholan
- Scientific Research Center, State Security Service Military Hospital, Baku, Azerbaijan
| | - Rufat Aliyev
- Scientific Research Center, State Security Service Military Hospital, Baku, Azerbaijan
| | - Ulduz Hashimova
- A. Karayev’s Institute of Physiology, Azerbaijan National Academy of Sciences, Baku, Azerbaijan
| | - Seymur Karimov
- Department of Kidney Diseases and Organ Transplantation, State Security Service Military Hospital, Baku, Azerbaijan
| | - Elvin Bayramov
- Department of Laboratory, State Security Service Military Hospital, Baku, Azerbaijan
| |
Collapse
|
3
|
Dika Ž, Živko M, Kljajić M, Jelaković B. SGLT2 Inhibitors and Their Effect on Urolithiasis: Current Evidence and Future Directions. J Clin Med 2024; 13:6017. [PMID: 39408078 PMCID: PMC11478155 DOI: 10.3390/jcm13196017] [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: 08/24/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Urolithiasis (UL) is increasingly prevalent due to rising cardiorenometabolic diseases, posing significant management challenges despite advances in urological techniques. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, primarily used for type 2 diabetes mellitus, chronic kidney disease, and heart failure, have emerged as a potential novel approach for UL treatment. These inhibitors may help reduce the risk of urolithiasis, particularly in patients with diabetes, by improving glycemic control and altering urinary chemistry, which are crucial factors in stone formation. However, the changes in urinary composition induced by SGLT2 inhibitors might also increase the risk of uric acid stone formation. This review evaluates the potential of SGLT2 inhibitors in managing UL, highlighting both the benefits and the risks. While these inhibitors show promise in reducing new and recurrent urinary stones in patients with diabetes, data on their effects in patients without diabetes who form stones are limited. Current human evidence largely comes from post hoc analyses of randomized controlled trials (RCTs) and large-scale database studies, with only one study providing detailed stone composition data. Experimental studies in animal models and cell lines have focused on calcium oxalate (CaOx) stones, showing that SGLT2 inhibitors specifically target CaOx stone formation and related renal inflammation. Although primarily studied for CaOx stones, their potential impact on other calcium-containing stones, such as calcium phosphate, remains promising. Further research is needed to explore their therapeutic potential and optimize treatment strategies.
Collapse
Affiliation(s)
- Živka Dika
- School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia;
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (M.Ž.); (M.K.)
| | - Marijana Živko
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (M.Ž.); (M.K.)
| | - Marina Kljajić
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (M.Ž.); (M.K.)
| | - Bojan Jelaković
- School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia;
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (M.Ž.); (M.K.)
| |
Collapse
|
4
|
Xu M, Qin Y, Xia Y, Wang G, Xiong Z, Song X, Ai L. Screening of oxalate-degrading probiotics and preventive effect of Lactiplantibacillus plantarum AR1089 on kidney stones. Food Funct 2024; 15:10163-10178. [PMID: 39300803 DOI: 10.1039/d4fo03133d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Calcium oxalate stone is the main type of kidney stone, so far there is no specific drug treatment. Here, we screened for oxalate-degrading probiotics and evaluated the potential preventive effect of lactic acid bacteria in rats with hyperoxaluria-induced kidney stones. The oxalate degradation efficiencies of the probiotics were determined to be 5-20% by in vitro experiments, of which the degradation efficiencies of Lactiplantibacillus plantarum AR342 and L. plantarum AR1089 were 17.32% and 14.15%, respectively. Through animal experiments, we found that L. plantarum AR1089 significantly attenuated kidney injury, as demonstrated by improving renal dysfunction and renal fibrosis, lowering creatinine and urea nitrogen levels. L. plantarum AR1089 was also effective in decreasing the number of calcium oxalate crystals in the urine and kidneys as well as ameliorating oxidative stress as evidenced by lowering the level of MDA and decreasing the level of SOD and CAT. Moreover, supplementation of L. plantarum AR1089 inhibited renal crystalline deposition by down-regulating the expression of KIM-1, OPN and MCP-1, and prevented hyperoxaluria-induced kidney stones by regulating the gut microbiota. Taken together, the present study shows that oral administration of L. plantarum AR1089, by attenuating kidney injury and regulating gut microbiota, is a potential therapy to reduce calcium oxalate crystals and prevent the progression of kidney stones.
Collapse
Affiliation(s)
- Mingyue Xu
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yugang Qin
- Aerospace Center Hospital, Beijing 100049, China
| | - Yongjun Xia
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Guangqiang Wang
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Zhiqiang Xiong
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Xin Song
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Lianzhong Ai
- Shanghai Engineering Research Center of Food Microbiology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
5
|
Muschialli L, Mannath A, Moochhala SH, Shroff R, Ferraro PM. Epidemiological and biological associations between cardiovascular disease and kidney stone formation: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis 2024; 34:559-568. [PMID: 38431384 DOI: 10.1016/j.numecd.2023.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 03/05/2024]
Abstract
AIMS Previous studies find kidney stone formers (KSF) are at greater risk of developing cardiovascular disease (CVD). The underlying mechanisms are poorly understood, and many clinicians are unaware of this connection. We will: DATA SYNTHESIS: Our systematic review is registered with PROSPERO (ID CRD42021251477). We searched epidemiological and biological data. The epidemiological search generated 669 papers, narrowed down to 15. There were 4,259,869 participants (230,720 KSFs). KSF was associated with 25% higher risk of coronary artery disease (CAD) (95% confidence interval (CI): 15, 35%), 17% higher risk of stroke/transient ischemic attacks (TIA) (CI:10, 25%) and 39% higher risk of arterial disease (AD) (CI: 17 65%). Significant heterogeneity was found. Female-identifying KSFs had a higher risk of stroke (ratio = 1.10) and CAD (1.20). The biological search generated 125 papers, narrowed down to 14. Potential underlying mechanisms were extracted and discussed, including intimal/medial vascular calcification, oxidative stress via osteopontin (OPN), cholesterol-induced pathology, and endothelial dysfunction. CONCLUSIONS There is a significant association between KSF and CVD, supporting the consideration of KSF as a systemic, calcium-mediated disease. Clinicians will benefit from being aware of this connection.
Collapse
Affiliation(s)
- Luke Muschialli
- UCL Medical School, Faculty of Medical Sciences, UCL, London, UK.
| | - Ankith Mannath
- UCL Medical School, Faculty of Medical Sciences, UCL, London, UK
| | | | - Rukshana Shroff
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
6
|
Huang L, Zhang Y, Fu H, Gu W, Mao J. A missense mutant of ocrl1 promotes apoptosis of tubular epithelial cells and disrupts endocytosis and the cell cycle of podocytes in Dent-2 Disease. Cell Commun Signal 2023; 21:256. [PMID: 38049819 PMCID: PMC10696739 DOI: 10.1186/s12964-023-01272-4] [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: 04/03/2023] [Accepted: 08/13/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND This study aimed to identify an orcl1 mutation in a patient with Dent-2 Disease and investigate the underlying mechanisms. METHODS The ocrl1 mutation was identified through exome sequencing. Knockdown of orcl1 and overexpression of the orcl1 mutant were performed in HK-2 and MPC5 cells to study its function, while flow cytometry measured reactive oxygen species (ROS), phosphatidylserine levels, and cell apoptosis. Scanning electron microscopy observed crystal adhesion, while transmission electron microscopy examined kidney tissue pathology. Laser scanning confocal microscopy was used to examine endocytosis, and immunohistochemical and immunofluorescence assays detected protein expression. Additionally, podocyte-specific orcl1 knockout mice were generated to investigate the role of orcl1 in vivo. RESULTS We identified a mutation resulting in the replacement of Histidine with Arginine at position 318 (R318H) in ocrl1 in the proband. orcl1 was widely expressed in the kidney. In vitro experiments showed that knockdown of orcl1 and overexpression of ocrl1 mutant increased ROS, phosphatidylserine exocytosis, crystal adhesion, and cell apoptosis in HK-2 cells. Knockdown of orcl1 in podocytes reduced endocytosis and disrupted the cell cycle while increasing cell migration. In vivo studies in mice showed that conditional deletion of orcl1 in podocytes caused glomerular dysfunction, including proteinuria and fibrosis. CONCLUSION This study identified an R318H mutation in orcl1 in a patient with Dent-2 Disease. This mutation may contribute to renal injury by promoting ROS production and inducing cell apoptosis in tubular cells, while disrupting endocytosis and the cell cycle, and promoting cell migration of podocytes. Video Abstract.
Collapse
Affiliation(s)
- Limin Huang
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine. National Clinical Research Center for Child Health, Hangzhou, China
| | - Yingying Zhang
- Department of Pediatrics, Clinical Center of Pediatric Nephrology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haidong Fu
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine. National Clinical Research Center for Child Health, Hangzhou, China
| | - Weizhong Gu
- Department of Pathologyology, Children's Hospital, Zhejiang University School of Medicine. National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine. National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
7
|
Kaneko K, Yasuda M, Fukuuchi T, Yamaoka N, Takahashi K, Mawatari KI, Isotani S, Horie S, Nakagawa T. Plasma levels and urinary excretion of protein Z in patients with urolithiasis. Int J Urol 2023; 30:1188-1193. [PMID: 37602692 DOI: 10.1111/iju.15277] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 08/02/2023] [Indexed: 08/22/2023]
Abstract
OBJECTIVES Protein Z (PZ) is a γ-carboxyglutamic acid protein present in plasma that is involved in blood coagulation. Detailed analysis of urinary stones from patients with urolithiasis has revealed that PZ is often found in urinary stones composed of calcium oxalate monohydrate. In this study, we compared blood and urinary PZ concentrations between healthy individuals and patients with urolithiasis. METHODS Plasma and urine were collected from healthy individuals and patients with urolithiasis who provided informed consent. PZ was detected as a urinary stone matrix protein in some of the patients. PZ was quantified by ELISA, creatinine was measured by the enzymatic method, and the total protein concentration was measured by the Bradford method. RESULTS The plasma PZ level was 2.54 ± 1.02 μg/mL in healthy individuals and that in urolithiasis patients classified by stone history were from 1.16 ± 0.77 to 3.73 ± 1.09 μg/mL, which was not significantly different. The urinary excretion of PZ (PZ/creatinine) was also not different in patients with urolithiasis and in healthy individuals (from 54.1 ± 40.9 to 95.4 ± 69.4 ng/mg vs. 73.3 ± 36.0 ng/mg). A positive correlation was found between the plasma PZ level and creatinine-corrected urinary PZ concentration (r = 0.46). CONCLUSIONS Both the plasma level and urinary excretion of PZ in urolithiasis patients were not significantly different with normal individuals. PZ detected in urinary stones as a matrix protein is thought to be incorporated into urinary stones regardless of blood and urine levels of PZ.
Collapse
Affiliation(s)
- Kiyoko Kaneko
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
- The Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma Sciences, Teikyo University, Tokyo, Japan
| | - Makoto Yasuda
- The Laboratory of Analytical Biochemistry, Faculty of Pharma Sciences, Teikyo University, Tokyo, Japan
| | - Tomoko Fukuuchi
- The Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma Sciences, Teikyo University, Tokyo, Japan
| | - Noriko Yamaoka
- The Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma Sciences, Teikyo University, Tokyo, Japan
| | - Kei Takahashi
- The Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma Sciences, Teikyo University, Tokyo, Japan
| | - Ken-Ichi Mawatari
- The Laboratory of Analytical Biochemistry, Faculty of Pharma Sciences, Teikyo University, Tokyo, Japan
| | - Shuji Isotani
- Department of Urology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigeo Horie
- Department of Urology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tohru Nakagawa
- Department of Urology, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Abstract
Oxalate homeostasis is maintained through a delicate balance between endogenous sources, exogenous supply and excretion from the body. Novel studies have shed light on the essential roles of metabolic pathways, the microbiome, epithelial oxalate transporters, and adequate oxalate excretion to maintain oxalate homeostasis. In patients with primary or secondary hyperoxaluria, nephrolithiasis, acute or chronic oxalate nephropathy, or chronic kidney disease irrespective of aetiology, one or more of these elements are disrupted. The consequent impairment in oxalate homeostasis can trigger localized and systemic inflammation, progressive kidney disease and cardiovascular complications, including sudden cardiac death. Although kidney replacement therapy is the standard method for controlling elevated plasma oxalate concentrations in patients with kidney failure requiring dialysis, more research is needed to define effective elimination strategies at earlier stages of kidney disease. Beyond well-known interventions (such as dietary modifications), novel therapeutics (such as small interfering RNA gene silencers, recombinant oxalate-degrading enzymes and oxalate-degrading bacterial strains) hold promise to improve the outlook of patients with oxalate-related diseases. In addition, experimental evidence suggests that anti-inflammatory medications might represent another approach to mitigating or resolving oxalate-induced conditions.
Collapse
Affiliation(s)
- Theresa Ermer
- Department of Surgery, Division of Thoracic Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Lama Nazzal
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Clarissa Tio
- Division of Nephrology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sushrut Waikar
- Department of Medicine, Section of Nephrology, Boston University, Boston, MA, USA
| | - Peter S Aronson
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Felix Knauf
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, USA.
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
9
|
Anan G, Hirose T, Kikuchi D, Takahashi C, Endo A, Ito H, Sato S, Nakayama S, Hashimoto H, Ishiyama K, Kimura T, Takahashi K, Sato M, Mori T. Inhibition of sodium-glucose cotransporter 2 suppresses renal stone formation. Pharmacol Res 2022; 186:106524. [PMID: 36349594 DOI: 10.1016/j.phrs.2022.106524] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND AIMS Nephrolithiasis is a common renal disease with no effective medication. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, an anti-diabetic agent, have diuretic and anti-inflammatory properties and could prevent nephrolithiasis. Here, we investigated the potential of SGLT2 inhibition against nephrolithiasis using large-scale epidemiological data, animal models, and cell culture experiments. METHODS This study included the data of diabetic patients (n = 1,538,198) available in the Japanese administrative database and divided them according to SGLT2 inhibitor prescription status. For animal experiments, renal calcium oxalate stones were induced by ethylene glycol in Sprague-Dawley rats, and phlorizin, an SGLT1/2 inhibitor, was used for the treatment. The effects of SGLT2-specific inhibition for renal stone formation were assessed in SGLT2-deficient mice and a human proximal tubular cell line, HK-2. RESULTS Nephrolithiasis prevalence in diabetic men was significantly lower in the SGLT2 inhibitor prescription group than in the non-SGLT2 inhibitor prescription group. Phlorizin attenuated renal stone formation and downregulated the kidney injury molecule 1 (Kim1) and osteopontin (Opn) expression in rats, with unchanged water intake and urine volume. It suppressed inflammation and macrophage marker expression, suggesting the role of the SGLT2 inhibitor in reducing inflammation. SGLT2-deficient mice were resistant to glyoxylic acid-induced calcium oxalate stone formation with reduced Opn expression and renal damages. High glucose-induced upregulation of OPN and CD44 and cell surface adhesion of calcium oxalate reduced upon SGLT2-silencing in HK-2 cells. CONCLUSION Overall, our findings identified that SGLT2 inhibition prevents renal stone formation and may be a promising therapeutic approach against nephrolithiasis.
Collapse
Affiliation(s)
- Go Anan
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Department of Urology, Yotsuya Medical Cube, Tokyo, Japan
| | - Takuo Hirose
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Division of Integrative Renal Replacement Therapy, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Daisuke Kikuchi
- Department of Pharmacy, Tohoku Medical and Pharmaceutical University Hospital, Sendai, Japan
| | - Chika Takahashi
- Division of Integrative Renal Replacement Therapy, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akari Endo
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroki Ito
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shigemitsu Sato
- Division of Integrative Renal Replacement Therapy, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shingo Nakayama
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hideaki Hashimoto
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Katsuya Ishiyama
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoyoshi Kimura
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Makoto Sato
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takefumi Mori
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Division of Integrative Renal Replacement Therapy, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
10
|
Yue L, Pai Q, Wu X, Zhang J. Smoking and Risk of Urolithiasis: Meta-Analysis of Observational Studies. Front Public Health 2022; 10:816756. [PMID: 35321192 PMCID: PMC8936389 DOI: 10.3389/fpubh.2022.816756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background Earlier studies have warned about the effects of smoking on urolithiasis. Some studies have deemed that smoking has a promoting effect on urolithiasis, whereas others have considered that no inevitable association exists between the two. Therefore, we conducted a meta-analysis to estimate whether smoking is associated with urolithiasis risk. Methods To identify publications from related observational studies, we performed a search on PubMed, Web of Science, Embase, and the Cochrane Library databases from inception until October 1, 2021. According to the heterogeneity, random-effect model was used to calculate the odds ratios (ORs) and corresponding 95% confidence intervals (CIs). Results Five articles were included in the meta-analysis, representing data for 20,402 subjects, of which 1,758 (8.62%) had urolithiasis as defined according to the criteria. Three articles are concerned with analysis between ex-smokers and non-smokers, in which a significant difference was observed (OR = 1.73, 95% CI: 1.48–2.01). Our comparison of current smokers with non-smokers in another meta-analysis of three articles revealed no significant difference between them (OR = 1.08, 95% CI: 0.94–1.23). Finally, we separated subjects into ever-smokers and never-smokers and found a significant difference between the two groups in the analysis of three articles (OR = 1.31, 95% CI: 1.17–1.47). Sensitivity analysis confirmed the stability of the current results. Conclusion Combined evidence from observational studies demonstrates a significant relation between smoking and urolithiasis. The trend of elevated urolithiasis risk from smoking was found in ever-smokers vs. never-smokers.
Collapse
|
11
|
Tanaka Y, Maruyama M, Okada A, Furukawa Y, Momma K, Sugiura Y, Tajiri R, Sawada KP, Tanaka S, Takano K, Taguchi K, Hamamoto S, Ando R, Tsukamoto K, Yoshimura M, Mori Y, Yasui T. Multicolor imaging of calcium-binding proteins in human kidney stones for elucidating the effects of proteins on crystal growth. Sci Rep 2021; 11:16841. [PMID: 34446727 PMCID: PMC8390759 DOI: 10.1038/s41598-021-95782-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/27/2021] [Indexed: 12/26/2022] Open
Abstract
The pathogenesis of kidney stone formation includes multi-step processes involving complex interactions between mineral components and protein matrix. Calcium-binding proteins in kidney stones have great influences on the stone formation. The spatial distributions of these proteins in kidney stones are essential for evaluating the in vivo effects of proteins on the stone formation, although the actual distribution of these proteins is still unclear. We reveal micro-scale distributions of three different proteins, namely osteopontin (OPN), renal prothrombin fragment 1 (RPTF-1), and calgranulin A (Cal-A), in human kidney stones retaining original mineral phases and textures: calcium oxalate monohydrate (COM) and calcium oxalate dihydrate (COD). OPN and RPTF-1 were distributed inside of both COM and COD crystals, whereas Cal-A was distributed outside of crystals. OPN and RPTF-1 showed homogeneous distributions in COM crystals with mosaic texture, and periodically distributions parallel to specific crystal faces in COD crystals. The unique distributions of these proteins enable us to interpret the different in vivo effects of each protein on CaOx crystal growth based on their physico-chemical properties and the complex physical environment changes of each protein. This method will further allow us to elucidate in vivo effects of different proteins on kidney stone formation.
Collapse
Affiliation(s)
- Yutaro Tanaka
- Department of Nephro-urology, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Mihoko Maruyama
- Institute for Advanced Co-Creation Studies, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Japan. .,Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Japan. .,Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5, Hangi-cho, Shimogamo, Sakyo-ku, Kyoto, Kyoto, 606-8522, Japan.
| | - Atsushi Okada
- Department of Nephro-urology, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-Ku, Nagoya, 467-8601, Japan.
| | - Yoshihiro Furukawa
- Department of Earth Science, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Koichi Momma
- National Museum of Nature and Science, 4-1-1 Amakubo, Tsukuba, 305-0005, Japan
| | - Yuki Sugiura
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa, 761-0395, Japan
| | - Rie Tajiri
- Tajiri Thin Section Laboratory, 3-1-11 Sannose, Higashiosaka, Osaka, 577-0849, Japan
| | - Koichi P Sawada
- Institute for Advanced Co-Creation Studies, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Japan
| | - Shunichi Tanaka
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5, Hangi-cho, Shimogamo, Sakyo-ku, Kyoto, Kyoto, 606-8522, Japan
| | - Kazufumi Takano
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5, Hangi-cho, Shimogamo, Sakyo-ku, Kyoto, Kyoto, 606-8522, Japan
| | - Kazumi Taguchi
- Department of Nephro-urology, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Ryosuke Ando
- Department of Nephro-urology, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Katsuo Tsukamoto
- Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Japan.,Department of Earth Science, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Masashi Yoshimura
- Institute of Laser Engineering, Osaka University, 2-6, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Yusuke Mori
- Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, 565-0871, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Graduate School of Medical Sciences, Nagoya City University, 1-Kawasumi, Mizuho-cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| |
Collapse
|
12
|
Randall's plaque and calcium oxalate stone formation: role for immunity and inflammation. Nat Rev Nephrol 2021; 17:417-433. [PMID: 33514941 DOI: 10.1038/s41581-020-00392-1] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 01/30/2023]
Abstract
Idiopathic calcium oxalate (CaOx) stones often develop attached to Randall's plaque present on kidney papillary surfaces. Similar to the plaques formed during vascular calcification, Randall's plaques consist of calcium phosphate crystals mixed with an organic matrix that is rich in proteins, such as inter-α-trypsin inhibitor, as well as lipids, and includes membrane-bound vesicles or exosomes, collagen fibres and other components of the extracellular matrix. Kidney tissue surrounding Randall's plaques is associated with the presence of classically activated, pro-inflammatory macrophages (also termed M1) and downregulation of alternatively activated, anti-inflammatory macrophages (also termed M2). In animal models, crystal deposition in the kidneys has been associated with the production of reactive oxygen species, inflammasome activation and increased expression of molecules implicated in the inflammatory cascade, including osteopontin, matrix Gla protein and fetuin A (also known as α2-HS-glycoprotein). Many of these molecules, including osteopontin and matrix Gla protein, are well known inhibitors of vascular calcification. We propose that conditions of urine supersaturation promote kidney damage by inducing the production of reactive oxygen species and oxidative stress, and that the ensuing inflammatory immune response promotes Randall's plaque initiation and calcium stone formation.
Collapse
|
13
|
Chaiyarit S, Thongboonkerd V. Mitochondrial Dysfunction and Kidney Stone Disease. Front Physiol 2020; 11:566506. [PMID: 33192563 PMCID: PMC7606861 DOI: 10.3389/fphys.2020.566506] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Mitochondrion is a pivotal intracellular organelle that plays crucial roles in regulation of energy production, oxidative stress, calcium homeostasis, and apoptosis. Kidney stone disease (nephrolithiasis/urolithiasis), particularly calcium oxalate (CaOx; the most common type), has been shown to be associated with oxidative stress and tissue inflammation/injury. Recent evidence has demonstrated the involvement of mitochondrial dysfunction in CaOx crystal retention and aggregation as well as Randall’s plaque formation, all of which are the essential mechanisms for kidney stone formation. This review highlights the important roles of mitochondria in renal cell functions and provides the data obtained from previous investigations of mitochondria related to kidney stone disease. In addition, mechanisms for the involvement of mitochondrial dysfunction in the pathophysiology of kidney stone disease are summarized. Finally, future perspectives on the novel approach to prevent kidney stone formation by mitochondrial preservation are discussed.
Collapse
Affiliation(s)
- Sakdithep Chaiyarit
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
14
|
Yamashita S, Komori T, Kohjimoto Y, Miyajima A, Hara I, Morikawa Y. Essential roles of oncostatin M receptor β signaling in renal crystal formation in mice. Sci Rep 2020; 10:17150. [PMID: 33051515 PMCID: PMC7553912 DOI: 10.1038/s41598-020-74198-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Oncostatin M (OSM), a member of the IL-6 family of cytokines, has important roles in renal diseases. The relationship between OSM and kidney stone disease, however, remains unclear. To investigate the roles of OSM in the development of kidney stone disease, we generated a mouse model of renal crystal formation using OSM receptor β (OSMRβ)-deficient mice (OSMRβ−/− mice). There were fewer renal crystal deposits in OSMRβ−/− mice than in wild-type (WT) mice. Crystal-binding molecules (osteopontin, annexin A1, and annexin A2), inflammatory cytokines (TNF-α and IL-1β), and fibrosis markers (TGF-β, collagen 1a2, and α-smooth muscle actin) were also decreased in the kidneys of OSMRβ−/− mice compared with those in WT mice. Immunofluorescence staining showed that OSMRβ was expressed in renal tubular epithelial cells (RTECs) and renal fibroblasts in the model of renal crystal formation. In the cultured RTECs and renal fibroblasts, OSM directly induced the expression of crystal-binding molecules and fibrosis markers. Expressions of inflammatory cytokines were increased by stimulation with OSM in cultured renal fibroblasts. OSM may promote the formation of renal crystal deposits by directly acting on RTECs and renal fibroblasts to produce crystal-binding molecules and inflammatory cytokines.
Collapse
Affiliation(s)
| | - Tadasuke Komori
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Yasuo Kohjimoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Yoshihiro Morikawa
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
15
|
Gabardo MCL, Kublitski PMDO, Sette IR, Lauschner T, Juglair MM, Baratto-Filho F, Brancher JA, Michel-Crosato E. Sialometric and Sialochemical Analysis in Individuals With Pulp Stones. Front Cell Dev Biol 2020; 8:403. [PMID: 32596238 PMCID: PMC7303886 DOI: 10.3389/fcell.2020.00403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/01/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to analyze the saliva of patients with pulp stones, with sialometric and sialochemical tests. Eighty individuals, aged between18 and 65 years, of both sexes, were investigated. Patients were included in the pulp stone group when radiographic examination was suggestive of pulp stones in at least one permanent tooth, whereas those without this alteration were considered controls. Saliva was collected by stimulation, followed by salivary flowrate (SFR) and pH analysis tests. The organic components, such as urea (URE), glucose (GLU), total proteins (TPTs), alkaline phosphatase, creatinine (CRE), salivary amylase (SAM), and uric acid (URA), and the inorganic components, such as calcium, iron, and phosphorus, were evaluated by colorimetric techniques in an ultraviolet–visible (UV–vis) spectrophotometer. Differences among pulp stones and control groups were compared using Student’s t-test, with a significance level of p < 0.05. In both groups prevailed the female. Statistically significant differences between groups were observed for pH (p = 0.027), SFR (p = 0.002), alkaline phosphatase (p = 0.008), and URA (p = 0.005). None of the inorganic components showed significant difference (p > 0.05). In the analyses stratified by sex, difference between groups was observed for pH (p = 0.007) and URA (p = 0.003) in women. In conclusion, sialometric and sialochemical alterations occurred in patients with pulp stones, with significantly higher levels of pH, SFR, alkaline phosphatase, and URA.
Collapse
Affiliation(s)
| | | | | | - Thaís Lauschner
- School of Health Sciences, Universidade Positivo, Curitiba, Brazil
| | | | | | | | | |
Collapse
|
16
|
Kuliasha CA, Rodriguez D, Lovett A, Gower LB. In situ flow cell platform for examining calcium oxalate and calcium phosphate crystallization on films of basement membrane extract in the presence of urinary 'inhibitors'. CrystEngComm 2020; 22:1448-1458. [PMID: 32256199 PMCID: PMC7111463 DOI: 10.1039/c9ce01587f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A significant portion of the population suffers from idipoathic calcium oxalate (CaOx) kidney stones, and current clinical treatments of stones have limited lasting success with a high rate of patients suffering from reoccurring stones. Understanding the role of physiologically relevant urinary species on the formation, aggregation, and growth of CaOx crystals can allow for better understanding of this complex biomineralization process and lead to more effective clinical treatments. Our prior work has focused on developing a two-stage model system, where the first stage emulates the formation of Randall's plaque, and the second stage examines the influence of the plaque on overgrowth of CaOx into a stone. Herein, we report on the development of an easy-to-use flow-cell platform that utilizes basement membrane extract (BME) as a biologically relevant crystallization substrate to study the influence of urinary 'inhibitors' on the in situ formation and growth of CaOx on BME under flow conditions. Magnesium, citrate, and osteopontin were studied because of their known ability to inhibit CaOx formation, but their influence also led to interesting modifications to the terminal crystal habit. Magnesium had little to no effect on the CaOx crystallization, but both citrate and osteopontin resulted in significant changes to the crystallization kinetics and the terminal crystal habits. Triply inhibited artificial urine solutions resulted in CaOx monohydrate formations that resembled physiological stones, and the in situ platform allowed for morphogenesis to be dynamically monitored. The BME was also used in a two-stage model system to first grow CaP that mimicked Randall's plaques, whereby the impact of the CaP crystallizing surface on CaOx formation could be studied. It was found that the CaP surface did not result in any significant changes in CaOx crystal formation or growth indicating that the urinary inhibitors and the basement membrane substrate were the dominant factors in modulating CaOx crystallization. It was also found that the basement membrane surface promoted the attachment and/or nucleation and growth of both CaOx and CaP crystals compared to bare glass surfaces, thereby enabling easy study of the urinary inhibitors. The work presented here has elucidated the terminal growth habit of different COM structures and has provided an easy to use platform that can be widely adopted by the kidney stone and other crystallization communities.
Collapse
Affiliation(s)
- Cary A. Kuliasha
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Douglas Rodriguez
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Archana Lovett
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Laurie B. Gower
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
17
|
Abstract
Infection stones are complex aggregates of crystals amalgamated in an organic matrix that are strictly associated with urinary tract infections. The management of patients who form infection stones is challenging owing to the complexity of the calculi and high recurrence rates. The formation of infection stones is a multifactorial process that can be driven by urine chemistry, the urine microenvironment, the presence of modulator substances in urine, associations with bacteria, and the development of biofilms. Despite decades of investigation, the mechanisms of infection stone formation are still poorly understood. A mechanistic understanding of the formation and growth of infection stones - including the role of organics in the stone matrix, microorganisms, and biofilms in stone formation and their effect on stone characteristics - and the medical implications of these insights might be crucial for the development of improved treatments. Tools and approaches used in various disciplines (for example, engineering, chemistry, mineralogy, and microbiology) can be applied to further understand the microorganism-mineral interactions that lead to infection stone formation. Thus, the use of integrated multidisciplinary approaches is imperative to improve the diagnosis, prevention, and treatment of infection stones.
Collapse
|
18
|
Unno R, Kawabata T, Taguchi K, Sugino T, Hamamoto S, Ando R, Okada A, Kohri K, Yoshimori T, Yasui T. Deregulated MTOR (mechanistic target of rapamycin kinase) is responsible for autophagy defects exacerbating kidney stone development. Autophagy 2019; 16:709-723. [PMID: 31257986 DOI: 10.1080/15548627.2019.1635382] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Kidney stone disease is a lifestyle-related disease prevalent in developed countries; however, effective medical treatment for the disease is not yet well established. As cellular damage in renal tubular cells (RTCs) is responsible for the disease, here, we focused on the role of macroautophagy/autophagy in RTCs. We found that autophagic activity was significantly decreased in mouse RTCs exposed to calcium oxalate (CaOx) monohydrate crystals and in the kidneys of GFP-conjugated MAP1LC3B (microtubule- associated protein 1 light chain 3 beta) transgenic mice with CaOx nephrocalcinosis induced by glyoxylate. This caused accumulation of damaged intracellular organelles, such as mitochondria and lysosomes, the normal functioning of which is mediated by functional autophagy. An impairment of autophagy was also observed in the mucosa with plaques of CaOx kidney stone formers. We determined that the decrease in autophagy was caused by an upregulation of MTOR (mechanistic target of rapamycin kinase), which consequently resulted in the suppression of the upstream autophagy regulator TFEB (transcription factor EB). Furthermore, we showed that an MTOR inhibitor could recover a decrease in autophagy and alleviate crystal-cell interactions and the formation of crystals associated with increased inflammatory responses. Taken together, we conclude that autophagy compromised by MTOR deregulation is a fundamental feature in the pathology of kidney stone formation, and propose that chemical inhibition of MTOR could be a prospective strategy for disease suppression.Abbreviations: ACTB: actin, beta; CaOx: calcium oxalate; CKD: chronic kidney disease; COM: calcium oxalate monohydrate; LGALS3/galectin-3: lectin, galactose binding, soluble 3; GFP: green fluorescent protein; GOX: glyoxylate; HE: hematoxylin and eosin; MAPLC3B: microtubule- associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; NAC: N-acetyl-L-cysteine; ROS: reactive oxygen species; RTC: renal tubular cell; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TEM: transmission electron microscopy; tfLC3: tandem fluorescent-tagged LC3; 3-MA: 3-methyladenine.
Collapse
Affiliation(s)
- Rei Unno
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Teruaki Sugino
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Ryosuke Ando
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Kenjiro Kohri
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| |
Collapse
|
19
|
Luukkonen J, Hilli M, Nakamura M, Ritamo I, Valmu L, Kauppinen K, Tuukkanen J, Lehenkari P. Osteoclasts secrete osteopontin into resorption lacunae during bone resorption. Histochem Cell Biol 2019; 151:475-487. [PMID: 30637455 PMCID: PMC6542781 DOI: 10.1007/s00418-019-01770-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2019] [Indexed: 01/27/2023]
Abstract
Osteopontin (OPN) is a non-collagenous extracellular sialylated glycoprotein located in bone. It is believed to be one of the key components in osteoclast attachment to bone during resorption. In this study, we characterized OPN and other glycoproteins found in the resorption lacunae to confirm the role of osteoclasts in OPN secretion using electron microscopy and mass spectrometry. Additionally, we examined the glycan epitopes of resorption pits and the effects of different glycan epitopes on the differentiation and function of osteoclasts. Osteoarthritic femoral heads were examined by immunohistochemistry to reveal the presence of OPN in areas of increased bone metabolism in vivo. Our results demonstrate that human osteoclasts secrete OPN into resorption lacunae on native human bone and on carbonated hydroxyapatite devoid of natural OPN. OPN is associated with an elevated bone turnover in osteoarthritic bone under experimental conditions. Our data further confirm that osteoclasts secrete OPN into the resorption pit where it may function as a chemokine for subsequent bone formation. We show that α2,3- and α2,6-linked sialic acids have a role in the process of osteoclast differentiation. OPN is one of the proteins that has both of the above sialic residues, hence we propose that de-sialylation can effect osteoclast differentiation in bone.
Collapse
Affiliation(s)
- Jani Luukkonen
- Department of Anatomy and Cell Biology, Cancer Research and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, Aapistie 5, 90014, Oulu, Finland.
| | - Meeri Hilli
- Department of Anatomy and Cell Biology, Cancer Research and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, Aapistie 5, 90014, Oulu, Finland
| | - Miho Nakamura
- Department of Anatomy and Cell Biology, Cancer Research and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, Aapistie 5, 90014, Oulu, Finland.,Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 1010062, Japan
| | - Ilja Ritamo
- Thermo Fisher Scientific Oy, Ratastie 2, 01620, Vantaa, Finland
| | - Leena Valmu
- Thermo Fisher Scientific Oy, Ratastie 2, 01620, Vantaa, Finland
| | - Kyösti Kauppinen
- Department of Anatomy and Cell Biology, Cancer Research and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, Aapistie 5, 90014, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Cancer Research and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, Aapistie 5, 90014, Oulu, Finland
| | - Petri Lehenkari
- Department of Anatomy and Cell Biology, Cancer Research and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, Aapistie 5, 90014, Oulu, Finland
| |
Collapse
|
20
|
Singhto N, Thongboonkerd V. Exosomes derived from calcium oxalate-exposed macrophages enhance IL-8 production from renal cells, neutrophil migration and crystal invasion through extracellular matrix. J Proteomics 2018; 185:64-76. [DOI: 10.1016/j.jprot.2018.06.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/06/2018] [Accepted: 06/18/2018] [Indexed: 12/11/2022]
|
21
|
Okada A, Hamamoto S, Taguchi K, Unno R, Sugino T, Ando R, Mizuno K, Tozawa K, Kohri K, Yasui T. Kidney stone formers have more renal parenchymal crystals than non-stone formers, particularly in the papilla region. BMC Urol 2018. [PMID: 29530009 PMCID: PMC5848581 DOI: 10.1186/s12894-018-0331-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We investigated the renoprotective ability of healthy people against kidney stone formation. To clarify intratubular crystal kinetics and processing in human kidneys, we performed a quantitative and morphological observation of nephrectomized renal parenchyma tissues. METHODS Clinical data and pathological samples from 60 patients who underwent radical nephrectomy for renal cancer were collected from June 2004 to June 2010. The patients were retrospectively classified as stone formers (SFs; n = 30, kidney stones detected by preoperative computed tomography) and non-stone formers (NSFs; n = 30, no kidney stone history). The morphology of parenchymal intratubular crystals and kidney stone-related gene and protein expression levels were examined in noncancerous renal sections from both groups. RESULTS SFs had a higher smoking rate (P = 0.0097); lower red blood cell, hemoglobin, and hematocrit values; and higher urinary red blood cell, white blood cell, and bacterial counts than NSFs. Scanning electron microscopy revealed calcium-containing crystal deposits and crystal attachment to the renal tubular lumen in both groups. Both groups demonstrated crystal transmigration from the tubular lumen to the interstitium. The crystal diffusion analysis indicated a significantly higher crystal existing ratio in the medulla and papilla of SFs and a significantly higher number of papillary crystal deposits in SFs than NSFs. The expression analysis indicated relatively high osteopontin and CD68, low superoxide dismutase, and significantly lower Tamm-Horsfall protein expression levels in SFs. Multivariate logistic regression analysis involving the above factors found the presence of renal papillary crystals as a significant independent factor related to SFs (odds ratio 5.55, 95% confidence interval 1.08-37.18, P = 0.0395). CONCLUSIONS Regardless of stone formation, intratubular crystals in the renal parenchyma seem to transmigrate to the interstitium. SFs may have reduced ability to eliminate renal parenchymal crystals, particularly those in the papilla region, than NSFs with associated gene expression profiles.
Collapse
Affiliation(s)
- Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan.
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Rei Unno
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Teruaki Sugino
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Ryosuke Ando
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Kentaro Mizuno
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Keiichi Tozawa
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Kenjiro Kohri
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| |
Collapse
|
22
|
Flanagan KC, Alspach E, Pazolli E, Parajuli S, Ren Q, Arthur LL, Tapia R, Stewart SA. c-Myb and C/EBPβ regulate OPN and other senescence-associated secretory phenotype factors. Oncotarget 2018; 9:21-36. [PMID: 29416593 PMCID: PMC5787458 DOI: 10.18632/oncotarget.22940] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023] Open
Abstract
Tumorigenesis results from the convergence of cell autonomous mutations and corresponding stromal changes that promote tumor cell growth. Senescent cells, which secrete a plethora of pro-tumorigenic factors termed the senescence-associated secretory phenotype (SASP), play an important role in tumor formation. Investigation into SASP regulation revealed that many but not all SASP factors are subject to NF-kB and p38MAPK regulation. However, many pro-tumorigenic SASP factors, including osteopontin (OPN), are not responsive to these canonical pathways leaving the regulation of these factors an open question. We report that the transcription factor c-Myb regulates OPN, IL-6, and IL-8 in addition to 57 other SASP factors. The regulation of OPN is direct as c-Myb binds to the OPN promoter in response to senescence. Further, OPN is also regulated by the known SASP regulator C/EBPβ. In response to senescence, the full-length activating C/EBPβ isoform LAP2 increases binding to the OPN, IL-6, and IL-8 promoters. The importance of both c-Myb and C/EBPβ is underscored by our finding that the depletion of either factor reduces the ability of senescent fibroblasts to promote the growth of preneoplastic epithelial cells.
Collapse
Affiliation(s)
- Kevin C. Flanagan
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- ICCE Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Elise Alspach
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ermira Pazolli
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shankar Parajuli
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qihao Ren
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura L. Arthur
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Roberto Tapia
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sheila A. Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- ICCE Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
Kim S, Chang Y, Sung E, Kang JG, Yun KE, Jung HS, Hyun YY, Lee KB, Joo KJ, Shin H, Ryu S. Association Between Sonographically Diagnosed Nephrolithiasis and Subclinical Coronary Artery Calcification in Adults. Am J Kidney Dis 2018; 71:35-41. [DOI: 10.1053/j.ajkd.2017.06.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/18/2017] [Indexed: 02/07/2023]
|
24
|
Joshi S, Wang W, Khan SR. Transcriptional study of hyperoxaluria and calcium oxalate nephrolithiasis in male rats: Inflammatory changes are mainly associated with crystal deposition. PLoS One 2017; 12:e0185009. [PMID: 29091707 PMCID: PMC5665423 DOI: 10.1371/journal.pone.0185009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
Hyperoxaluria associated with renal deposition of calcium oxalate (CaOx) crystals causes renal injury and inflammation leading to number of diseases including chronic kidney disease (CKD). It is however, not been possible to separate the renal consequences of hyperoxaluria from that of CaOx crystal deposition. We decided to utilize ethylene glycol (EG) model where hyperoxaluria and CaOx crystal deposition can be separated in time. To test our hypothesis, male rats were made hyperoxaluric by administering EG, rats were euthanized and kidneys were extracted on day 14, when occasional crystal is seen in the kidneys and day 28, when all animals have developed renal CaOx crystal deposits. Total RNA was extracted for microarray analysis and genome wide analysis of differentially expressed genes was performed to investigate differences between hyperoxaluria and crystal induced alterations in the kidneys. Immunohistochemical and Hematoxylin and Eosin (H&E) staining was also done for macromolecules with significant role in stone formation. All EG fed rats became hyperoxaluric by day 7, showed a few crystal deposits on day 14, and had heavy crystal deposition by day 28. There were significant changes in the expression of genes encoding for NADPH Oxidases; macromolecular crystallization modulators; genes involved in inflammasome activation; and osteogenic marker genes. Results demonstrate major differences between hyperoxaluria and CaOx crystal induced changes in the kidneys. Injury and inflammation are mainly associated with crystal deposition indicating significant role played by crystal retention.
Collapse
Affiliation(s)
- Sunil Joshi
- Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Wei Wang
- Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Saeed R. Khan
- Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Urology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
25
|
Taguchi K, Yasui T, Milliner DS, Hoppe B, Chi T. Genetic Risk Factors for Idiopathic Urolithiasis: A Systematic Review of the Literature and Causal Network Analysis. Eur Urol Focus 2017; 3:72-81. [PMID: 28720371 DOI: 10.1016/j.euf.2017.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/29/2017] [Indexed: 02/06/2023]
Abstract
CONTEXT Urolithiasis has a high prevalence and recurrence rate. Prevention is key to patient management, but risk stratification is challenging. In particular, genetic predisposition for urinary stones is not fully understood. OBJECTIVE To review current evidence of potential causative genes for idiopathic urolithiasis and map their relationships to one another. This evidence is essential for future establishment of molecular targeted therapy. EVIDENCE ACQUISITION A systematic literature review from 2007 to 2017 was performed in accordance with the Preferred Reporting Items for Systematic Review and Meta-analyses guidelines. The search was restricted to human studies conducted as either case-control or genome-wide association studies, and published in English. We also performed a causal network analysis of candidate genes gained from the systematic review using Ingenuity Pathway Analysis (IPA). EVIDENCE SYNTHESIS During the systematic screening of literature, 30 papers were selected for the review. A total of 20 genes with 42 polymorphisms/variants were found to be associated with urolithiasis risk. Their functional roles were mainly categorized as stone matrix, calcium and phosphate regulation, urinary concentration and constitution, and inflammation/oxidative stress. IPA network analysis revealed that these genes connected via signaling pathways and a proinflammatory/oxidative environment. CONCLUSIONS This systematic review provides an updated gene list and novel causal networks for idiopathic urolithiasis risk. Although some genes such as SPP1, CASR, VDR, CLDN14, and SLC34A1 were identified by several studies and recognized by prior reviews, further investigation elucidating their roles in stone formation will be essential for future studies. PATIENT SUMMARY In this review, we summarized recent literature regarding genes responsible for kidney stone risk. Based on a detailed review of 30 articles and computational network analysis, we concluded that disorder of mineral regulation with local inflammation in the kidney may cause kidney stone disease.
Collapse
Affiliation(s)
- Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Department of Urology, University of California, San Francisco, CA, USA
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Dawn Schmautz Milliner
- Division of Nephrology, Departments of Pediatrics and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Bernd Hoppe
- Division of Pediatric Nephrology, Department of Pediatrics, University Hospital Bonn, Bonn, Germany
| | - Thomas Chi
- Department of Urology, University of California, San Francisco, CA, USA.
| |
Collapse
|
26
|
Lee SY, Lee SJ, Piao HL, Yang SY, Weiner ID, Kim J, Han KH. Hydration status affects osteopontin expression in the rat kidney. J Vet Sci 2017; 17:269-77. [PMID: 26645343 PMCID: PMC5037293 DOI: 10.4142/jvs.2016.17.3.269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/13/2015] [Accepted: 10/07/2015] [Indexed: 12/22/2022] Open
Abstract
Osteopontin (OPN) is a secretory protein that plays an important role in urinary stone formation. Hydration status is associated with the development of urolithiasis. This study was conducted to examine the effects of dehydration and hydration on OPN expression in the rat kidney. Animals were divided into three groups, control, dehydrated, and hydrated. Kidney tissues were processed for light and electron microscope immunocytochemistry, in situ hybridization, and immunoblot analysis. Dehydration induced a significant increase in OPN protein expression, whereas increased fluid intake induced a decrease in protein expression. Under control conditions, OPN protein and mRNA expression were only detected in the descending thin limb (DTL). Dehydration induced increased expression in the DTL and the development of detectable expression in the thick ascending limb (TAL). In contrast, OPN expression levels declined to less than the controls in the DTL after hydration, while no expression of either protein or mRNA was detectable in the TAL. Immunoelectron microscopy demonstrated that hydration status altered tubular ultrastructure and intracellular OPN expression in the Golgi apparatus and secretory cytoplasmic vesicles. These data confirm that changes in oral fluid intake can regulate renal tubular epithelial cell OPN expression.
Collapse
Affiliation(s)
- Su-Youn Lee
- Department of Anatomy, Ewha Womans University School of Medicine, Seoul 03760, Korea
| | - Sae-Jin Lee
- Department of Anatomy, Ewha Womans University School of Medicine, Seoul 03760, Korea
| | - Hong-Lin Piao
- Department of Anatomy, Ewha Womans University School of Medicine, Seoul 03760, Korea
| | - Suk-Young Yang
- Department of Anatomy, Ewha Womans University School of Medicine, Seoul 03760, Korea
| | - I David Weiner
- Division of Nephrology, College of Medicine, University of Florida, Gainesville, FL 32608, USA.,Nephrology Section, North Florida/South Georgia Veterans Health System (NF/SGVHS), Gainesville, FL 32608, USA
| | - Jin Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Hwan Han
- Department of Anatomy, Ewha Womans University School of Medicine, Seoul 03760, Korea
| |
Collapse
|
27
|
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
|
28
|
Khan SR. Histological aspects of the "fixed-particle" model of stone formation: animal studies. Urolithiasis 2016; 45:75-87. [PMID: 27896391 DOI: 10.1007/s00240-016-0949-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/22/2016] [Indexed: 01/25/2023]
Abstract
Crystallization by itself is not harmful as long as the crystals are not retained in the kidneys and are allowed to pass freely down the renal tubules to be excreted in the urine. A number of theories have been proposed, and studies performed, to determine the mechanisms involved in crystal retention within the kidneys. It has been suggested that urinary transit through the nephron is too fast for crystals to grow large enough to be retained. Thus, free particle mechanism alone cannot lead to stone formation, and there must be a mechanism for crystal fixation within the kidneys. Animal model studies suggest that crystal retention is possible through both the free- and fixed-particle mechanisms. Crystal-cell interaction leads to pathological changes which promote crystal attachment to either epithelial cells or their basement membrane. Alternatively, crystals aggregate and produce large enough particles to block the tubules particularly at sites, where urinary flow is affected because of changes in the luminal diameter of the tubule. Crystal deposits plugging the openings of the ducts of Bellini may be the result of such a phenomenon. Intratubular crystals translocating to renal interstitium may produce osteogenic changes in the epithelial or endothelial cells resulting in the formation of the Randall's plaques. Thus, fixation appears to be either through the formation of Randall's plugs, crystal plugs clogging the openings of the ducts of Bellini or sub-epithelial crystal deposits, and the Randall's plaques.
Collapse
Affiliation(s)
- Saeed R Khan
- Department of Pathology, Immunology and Laboratory Investigation, College of Medicine, University of Florida, Gainesville, FL, USA. .,Department of Urology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
29
|
M1/M2-macrophage phenotypes regulate renal calcium oxalate crystal development. Sci Rep 2016; 6:35167. [PMID: 27731368 PMCID: PMC5059697 DOI: 10.1038/srep35167] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/20/2016] [Indexed: 12/28/2022] Open
Abstract
In our previous report, M2-macrophage (Mφs) deficient mice showed increased renal calcium oxalate (CaOx) crystal formation; however, the role of Mφs-related-cytokines and chemokines that affect kidney stone formation remains unknown. Here, we investigated the role of M1/M2s in crystal development by using in vitro and in vivo approaches. The crystal phagocytic rate of bone marrow-derived M2Mφs was higher than that of bone marrow-derived Mφs and M1Mφs and increased on co-culture with renal tubular cells (RTCs). However, the amount of crystal attachment on RTCs reduced on co-culture with M2Mφs. In six hyperoxaluric C57BL/6J mice, M1Mφ transfusion and induction by LPS and IFN-γ facilitated renal crystal formation, whereas M2Mφ transfusion and induction by IL-4 and IL-13 suppressed renal crystal formation compared with the control. These M2Mφ treatments reduced the expression of crystal-related genes, such as osteopontin and CD44, whereas M1Mφ treatment increased the expression of pro-inflammatory and adhesion-related genes such as IL-6, inducible NOS, TNF-α, C3, and VCAM-1. The expression of M2Mφ-related genes was lower whereas that of M1Mφ-related genes was higher in papillary tissue of CaOx stone formers. Overall, our results suggest that renal crystal development is facilitated by M1Mφs, but suppressed by M2Mφs.
Collapse
|
30
|
Yasui T, Okada A, Hamamoto S, Ando R, Taguchi K, Tozawa K, Kohri K. Pathophysiology-based treatment of urolithiasis. Int J Urol 2016; 24:32-38. [DOI: 10.1111/iju.13187] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/18/2016] [Indexed: 12/01/2022]
Affiliation(s)
- Takahiro Yasui
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Atsushi Okada
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Ryosuke Ando
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Kazumi Taguchi
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Keiichi Tozawa
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Kenjiro Kohri
- Department of Nephro-urology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| |
Collapse
|
31
|
Taguchi K, Hamamoto S, Okada A, Unno R, Kamisawa H, Naiki T, Ando R, Mizuno K, Kawai N, Tozawa K, Kohri K, Yasui T. Genome-Wide Gene Expression Profiling of Randall's Plaques in Calcium Oxalate Stone Formers. J Am Soc Nephrol 2016; 28:333-347. [PMID: 27297950 DOI: 10.1681/asn.2015111271] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/04/2016] [Indexed: 12/23/2022] Open
Abstract
Randall plaques (RPs) can contribute to the formation of idiopathic calcium oxalate (CaOx) kidney stones; however, genes related to RP formation have not been identified. We previously reported the potential therapeutic role of osteopontin (OPN) and macrophages in CaOx kidney stone formation, discovered using genome-recombined mice and genome-wide analyses. Here, to characterize the genetic pathogenesis of RPs, we used microarrays and immunohistology to compare gene expression among renal papillary RP and non-RP tissues of 23 CaOx stone formers (SFs) (age- and sex-matched) and normal papillary tissue of seven controls. Transmission electron microscopy showed OPN and collagen expression inside and around RPs, respectively. Cluster analysis revealed that the papillary gene expression of CaOx SFs differed significantly from that of controls. Disease and function analysis of gene expression revealed activation of cellular hyperpolarization, reproductive development, and molecular transport in papillary tissue from RPs and non-RP regions of CaOx SFs. Compared with non-RP tissue, RP tissue showed upregulation (˃2-fold) of LCN2, IL11, PTGS1, GPX3, and MMD and downregulation (0.5-fold) of SLC12A1 and NALCN (P<0.01). In network and toxicity analyses, these genes associated with activated mitogen-activated protein kinase, the Akt/phosphatidylinositol 3-kinase pathway, and proinflammatory cytokines that cause renal injury and oxidative stress. Additionally, expression of proinflammatory cytokines, numbers of immune cells, and cellular apoptosis increased in RP tissue. This study establishes an association between genes related to renal dysfunction, proinflammation, oxidative stress, and ion transport and RP development in CaOx SFs.
Collapse
Affiliation(s)
- Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and.,Department of Urology, Social Medical Corporation Kojunkai Daido Hospital, Daido Clinic, Nagoya, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Rei Unno
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Hideyuki Kamisawa
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and.,Department of Urology, Social Medical Corporation Kojunkai Daido Hospital, Daido Clinic, Nagoya, Japan
| | - Taku Naiki
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Ryosuke Ando
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Kentaro Mizuno
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Noriyasu Kawai
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Keiichi Tozawa
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Kenjiro Kohri
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; and
| |
Collapse
|
32
|
Christensen B, Zachariae ED, Scavenius C, Kløverpris S, Oxvig C, Petersen SV, Enghild JJ, Sørensen ES. Transglutaminase 2-Catalyzed Intramolecular Cross-Linking of Osteopontin. Biochemistry 2016; 55:294-303. [PMID: 26678563 DOI: 10.1021/acs.biochem.5b01153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Osteopontin (OPN) is a multifunctional integrin-binding protein present in several tissues and body fluids. OPN is a substrate for the enzyme transglutaminase 2 (TG2), which catalyzes inter- and intramolecular cross-linking affecting the biological activity of the protein. Polymerization of OPN by intermolecular cross-linking has mostly been studied using relatively high TG2 concentrations, whereas the effect of lower concentrations of TG2 has remained unexplored. Here we show that TG2 at physiologically relevant concentrations predominantly catalyzes the formation of intramolecular cross-links in OPN. By site-directed mutagenesis and mass spectrometry, we demonstrate that Gln(42) and Gln(193) serve as the primary amine acceptor sites for isopeptide bond formation. We find that Gln(42) predominantly is linked to Lys(4) and that Gln(193) participates in a cross-link with Lys(154), Lys(157), or Lys(231). The formation of specific isopeptide bonds was not dependent on OPN phosphorylation, and similar patterns of cross-linking were observed in human and mouse OPN. Furthermore, we find that OPN purified from human urine contains the Lys(154)-Gln(193) isopeptide bond, indicating that intramolecular cross-linking of OPN occurs in vivo. Collectively, these data suggest that specific intramolecular cross-linking in the N- and C-terminal parts of OPN is most likely the dominant step in TG2-catalyzed modification of OPN.
Collapse
Affiliation(s)
- Brian Christensen
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Elias D Zachariae
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Søren Kløverpris
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Steen V Petersen
- Department of Biomedicine, Aarhus University , 8000 Aarhus C, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University , 8000 Aarhus C, Denmark
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University , 8000 Aarhus C, Denmark
| |
Collapse
|
33
|
Liu Y, Xu H, Zhong W, Shen Q, Zhuang T, Huang K. Organic Selenium Alleviated the Formation of Ethylene Glycol-Induced Calcium Oxalate Renal Calculi by Improving Osteopontin Expression and Antioxidant Capability in Dogs. Biol Trace Elem Res 2015; 168:392-400. [PMID: 26018495 DOI: 10.1007/s12011-015-0373-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/14/2015] [Indexed: 10/23/2022]
Abstract
Twenty one-year-old local male dogs were randomly assigned into four groups (five dogs per group). The control and the ethylene glycol (EG) groups were fed basal diets without and with EG, and the EG+sodium selenite (EG+SS) and EG+selenium yeast (EG+SY) groups were fed basal diets with EG containing SS and SY, respectively. Blood, urine, and renal samples were taken after 18 weeks of feeding. The results showed that compared with the control group, the serum calcium levels and antioxidase activities significantly decreased in the EG group. Serum creatinine, urea nitrogen, and malondialdehyde (MDA) levels and urine calcium and oxalate levels significantly increased. Calcium oxalate crystal deposition and osteopontin (OPN) messenger RNA and protein expression in the renal tissues significantly increased. These changes above in the EG group were reversed within limits by adding selenium in the diets (both EG+SS and EG+SY groups). Further, compared with the EG+SS group, the EG+SY group showed better effects in decreasing the formation of EG-induced calcium oxalate renal calculi and OPN expression and improving antioxidant capability in dogs. It indicates that organic selenium has the potential value to alleviate the formation of EG-induced calcium oxalate renal calculi.
Collapse
Affiliation(s)
- Yongwang Liu
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, China
| | - Haibin Xu
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wenting Zhong
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qingpeng Shen
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tenghan Zhuang
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, China
| | - Kehe Huang
- Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
34
|
Proinflammatory and Metabolic Changes Facilitate Renal Crystal Deposition in an Obese Mouse Model of Metabolic Syndrome. J Urol 2015; 194:1787-96. [PMID: 26192255 DOI: 10.1016/j.juro.2015.07.083] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2015] [Indexed: 01/19/2023]
Abstract
PURPOSE To clarify metabolic syndrome induced stone formation mechanisms we investigated the metabolic and immunohistochemical characteristics associated with renal crystal deposition using a model of mice with metabolic syndrome administered a high fat diet and ethylene glycol. MATERIALS AND METHODS Ob/Ob mice with Leptin gene deficiencies and metabolic syndrome related characteristics were compared with wild heterozygous lean mice. Four study groups were fed standard food and water (control group), a high fat diet and normal water (high fat diet group), 1% ethylene glycol and standard food (ethylene glycol group) or a high fat diet and 1% ethylene glycol (high fat diet plus ethylene glycol group). Blood, urine and kidney samples were taken after 14 days. RESULTS Ob/Ob mice in the high fat diet plus ethylene glycol group showed diffuse renal crystal depositions. Lean and Ob/Ob mice in the high fat diet plus ethylene glycol group showed significant excretion of urinary calcium oxalate crystals. Ob/Ob mice had significant hypercalciuria, hyperphosphaturia and hyperlipidemia, massive lipid fragments in tubular lumina and fat droplets in renal tubular cells. Ob/Ob mice in the high fat diet plus ethylene glycol group had markedly increased expression of osteopontin, monocyte chemoattractant protein-1, interleukin-6 and tumor necrosis factor-α. In Ob/Ob mice the number of proinflammatory macrophages was considerably elevated. CONCLUSIONS We induced renal crystal deposition in mice with metabolic syndrome using a high fat diet and ethylene glycol. Increases in luminal mineral and lipid density, and proinflammatory adipocytokines and macrophages facilitated renal crystal formation in mice with metabolic syndrome.
Collapse
|
35
|
Kläning E, Christensen B, Sørensen ES, Vorup-Jensen T, Jensen JK. Osteopontin binds multiple calcium ions with high affinity and independently of phosphorylation status. Bone 2014; 66:90-5. [PMID: 24928493 DOI: 10.1016/j.bone.2014.05.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/15/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
Osteopontin (OPN) is an acidic, intrinsically disordered extracellular matrix protein with a capacity to modulate biomineralization in vitro and in vivo. The role of posttranslational modification of osteopontin has been intensively studied. Phosphorylation of OPN has been demonstrated to play a role in inhibition of biomineral formation and growth in vitro. Here, we used isothermal titration calorimetry (ITC) to investigate the ability of OPN to bind the divalent cations Ca(2+) and Mg(2+), both essential components of inorganic minerals in vivo. We found, that bovine OPN binds ~10 Ca(2+) ions with an apparent affinity ~50-fold tighter than Mg(2+), both regardless of OPN phosphorylation, and with affinities significantly stronger than previously reported. These results were confirmed using human derived OPN. This implies that a majority of the acidic residues within OPN must be engaged in calcium interaction under physiological conditions.
Collapse
Affiliation(s)
- Eva Kläning
- Department of Molecular Biology and Genetics, Aarhus University, Denmark; Department of Biomedicine, Aarhus University, Denmark
| | - Brian Christensen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | | | - Jan K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark; Danish-Chinese Centre for Proteases and Cancer, Aarhus University, Denmark.
| |
Collapse
|
36
|
Khan SR, Canales BK. Unified theory on the pathogenesis of Randall's plaques and plugs. Urolithiasis 2014; 43 Suppl 1:109-23. [PMID: 25119506 DOI: 10.1007/s00240-014-0705-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/23/2014] [Indexed: 01/19/2023]
Abstract
Kidney stones develop attached to sub-epithelial plaques of calcium phosphate (CaP) crystals (termed Randall's plaque) and/or form as a result of occlusion of the openings of the Ducts of Bellini by stone-forming crystals (Randall's plugs). These plaques and plugs eventually extrude into the urinary space, acting as a nidus for crystal overgrowth and stone formation. To better understand these regulatory mechanisms and the pathophysiology of idiopathic calcium stone disease, this review provides in-depth descriptions of the morphology and potential origins of these plaques and plugs, summarizes existing animal models of renal papillary interstitial deposits, and describes factors that are believed to regulate plaque formation and calcium overgrowth. Based on evidence provided within this review and from the vascular calcification literature, we propose a "unified" theory of plaque formation-one similar to pathological biomineralization observed elsewhere in the body. Abnormal urinary conditions (hypercalciuria, hyperoxaluria, and hypocitraturia), renal stress or trauma, and perhaps even the normal aging process lead to transformation of renal epithelial cells into an osteoblastic phenotype. With this de-differentiation comes an increased production of bone-specific proteins (i.e., osteopontin), a reduction in crystallization inhibitors (such as fetuin and matrix Gla protein), and creation of matrix vesicles, which support nucleation of CaP crystals. These small deposits promote aggregation and calcification of surrounding collagen. Mineralization continues by calcification of membranous cellular degradation products and other fibers until the plaque reaches the papillary epithelium. Through the activity of matrix metalloproteinases or perhaps by brute physical force produced by the large sub-epithelial crystalline mass, the surface is breached and further stone growth occurs by organic matrix-associated nucleation of CaOx or by the transformation of the outer layer of CaP crystals into CaOx crystals. Should this theory hold true, developing an understanding of the cellular mechanisms involved in progression of a small, basic interstitial plaque to that of an expanding, penetrating plaque could assist in the development of new therapies for stone prevention.
Collapse
Affiliation(s)
- Saeed R Khan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA,
| | | |
Collapse
|
37
|
Interstitial calcinosis in renal papillae of genetically engineered mouse models: relation to Randall's plaques. Urolithiasis 2014; 43 Suppl 1:65-76. [PMID: 25096800 DOI: 10.1007/s00240-014-0699-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 07/22/2014] [Indexed: 02/08/2023]
Abstract
Genetically engineered mouse models (GEMMs) have been highly instrumental in elucidating gene functions and molecular pathogenesis of human diseases, although their use in studying kidney stone formation or nephrolithiasis remains relatively limited. This review intends to provide an overview of several knockout mouse models that develop interstitial calcinosis in the renal papillae. Included herein are mice deficient for Tamm-Horsfall protein (THP; also named uromodulin), osteopontin (OPN), both THP and OPN, Na(+)-phosphate cotransporter Type II (Npt2a) and Na(+)/H(+) exchanger regulatory factor (NHERF-1). The baseline information of each protein is summarized, along with key morphological features of the interstitial calcium deposits in mice lacking these proteins. Attempts are made to correlate the papillary interstitial deposits found in GEMMs with Randall's plaques, the latter considered precursors of idiopathic calcium stones in patients. The pathophysiology that underlies the renal calcinosis in the knockout mice is also discussed wherever information is available. Not all the knockout models are allocated equal space because some are more extensively characterized than others. Despite the inroads already made, the exact physiological underpinning, origin, evolution and fate of the papillary interstitial calcinosis in the GEMMs remain incompletely defined. Greater investigative efforts are warranted to pin down the precise role of the papillary interstitial calcinosis in nephrolithiasis using the existing models. Additionally, more sophisticated, second-generation GEMMs that allow gene inactivation in a time-controlled manner and "compound mice" that bear several genetic alterations are urgently needed, in light of mounting evidence that nephrolithiasis is a multifactorial, multi-stage and polygenic disease.
Collapse
|
38
|
Osteopontin knockdown in the kidneys of hyperoxaluric rats leads to reduction in renal calcium oxalate crystal deposition. Urolithiasis 2014; 42:195-202. [PMID: 24619192 DOI: 10.1007/s00240-014-0649-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 01/16/2014] [Indexed: 12/26/2022]
Abstract
Osteopontin (OPN) expression is increased in kidneys of rats with ethylene glycol (EG) induced hyperoxaluria and calcium oxalate (CaOx) nephrolithiasis. The aim of this study is to clarify the effect of OPN knockdown by in vivo transfection of OPN siRNA on deposition of CaOx crystals in the kidneys. Hyperoxaluria was induced in 6-week-old male Sprague-Dawley rats by administering 1.5% EG in drinking water for 2 weeks. Four groups of six rats each were studied: Group A, untreated animals (tap water); Group B, administering 1.5% EG; Group C, 1.5% EG with in vivo transfection of OPN siRNA; Group D, 1.5% EG with in vivo transfection of negative control siRNA. OPN siRNA transfections were performed on day 1 and 8 by renal sub-capsular injection. Rats were killed at day 15 and kidneys were removed. Extent of crystal deposition was determined by measuring renal calcium concentrations and counting renal crystal deposits. OPN siRNA transfection resulted in significant reduction in expression of OPN mRNA as well as protein in group C compared to group B. Reduction in OPN expression was associated with significant decrease in crystal deposition in group C compared to group B. Specific suppression of OPN mRNA expression in kidneys of hyperoxaluric rats leads to a decrease in OPN production and simultaneously inhibits renal crystal deposition.
Collapse
|
39
|
Khan SR, Joshi S, Wang W, Peck AB. Regulation of macromolecular modulators of urinary stone formation by reactive oxygen species: transcriptional study in an animal model of hyperoxaluria. Am J Physiol Renal Physiol 2014; 306:F1285-95. [PMID: 24598804 DOI: 10.1152/ajprenal.00057.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We used an unbiased approach of gene expression profiling to determine differential gene expression of all the macromolecular modulators (MMs) considered to be involved in stone formation, in hyperoxaluric rats, with and without treatment with the NADPH oxidase inhibitor apocynin. Male rats were fed rat chow or chow supplemented with 5% wt/wt hydroxy-l-proline (HLP) with or without apocynin-supplemented water. After 28 days, rats were euthanized and their kidneys explanted. Total RNA was isolated and microarray analysis was conducted using the Illumina bead array reader. Gene ontology analysis and the pathway analyses of the genes were done using Database for Annotation, Visualization of Integrated Discovery enrichment analysis tool. Quantitative RT-PCR of selected genes was carried out to verify the microarray results. Expression of selected gene products was confirmed using immunohistochemistry. Administration of HLP led to crystal deposition. Genes encoding for fibronectin, CD 44, fetuin B, osteopontin, and matrix-gla protein were upregulated while those encoding for heavy chains of inter-alpha-inhibitor 1, 3, and 4, calgranulin B, prothrombin, and Tamm-Horsfall protein were downregulated. HLP-fed rats receiving apocynin had a significant reversal in gene expression profiles: those that were upregulated came down while those that were downregulated stepped up. Apocynin treatment resulted in near complete absence of crystals. Clearly, there are two types of MMs; one is downregulated while the other is upregulated during hyperoxaluria and crystal deposition. Apparently gene and protein expressions of known macromolecular modulators of CaOx crystallization are likely regulated by ROS produced in part through the activation of NADPH oxidase.
Collapse
Affiliation(s)
- Saeed R Khan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida; Department of Urology, College of Medicine, University of Florida, Gainesville, Florida; and
| | - Sunil Joshi
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Wei Wang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Ammon B Peck
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
40
|
Vlasakova K, Erdos Z, Troth SP, McNulty K, Chapeau-Campredon V, Mokrzycki N, Muniappa N, Gu YZ, Holder D, Bailey WJ, Sistare FD, Glaab WE. Evaluation of the Relative Performance of 12 Urinary Biomarkers for Renal Safety Across 22 Rat Sensitivity and Specificity Studies. Toxicol Sci 2013; 138:3-20. [DOI: 10.1093/toxsci/kft330] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
41
|
Green W, Ratan H. Molecular mechanisms of urolithiasis. Urology 2013; 81:701-4. [PMID: 23434095 DOI: 10.1016/j.urology.2012.12.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/05/2012] [Accepted: 12/28/2012] [Indexed: 10/27/2022]
Affiliation(s)
- Will Green
- Department of Urology, Royal Derby Hospital, Derby, United Kingdom.
| | | |
Collapse
|