1
|
Gurevich E, Landau D. Tubulointerstitial nephritis in children and adolescents. Pediatr Nephrol 2025; 40:319-328. [PMID: 39320551 DOI: 10.1007/s00467-024-06526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
The tubulointerstitial compartment comprises most of the kidney parenchyma. Inflammation in this compartment (tubulointerstitial nephritis-TIN) can be acute and resolves if the offending factor is withdrawn or may enter a chronic process leading to irreversible kidney damage. Etiologic factors differ, including different exposures, infections, and autoimmune and genetic tendency, and the initial damage can be acute, recurrent, or permanent, determining whether the acute inflammatory process will lead to complete healing or to a chronic course of inflammation leading to fibrosis. Clinical and laboratory findings of TIN are often nonspecific, which may lead to delayed diagnosis and a poorer clinical outcome. We provide a general review of TIN, with special mention of the molecular pathophysiological mechanisms of the associated kidney damage.
Collapse
Affiliation(s)
- Evgenia Gurevich
- Pediatrics Department, Barzilai University Medical Center, Ashqelon, Israel.
- Ben Gurion University of Negev, Faculty of Health Sciences, Beer Sheva, Israel.
| | - Daniel Landau
- Department of Nephrology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Levi S, Landau D, Davidovits M, Shapira Rootman M, Brener A, Gal S, Borovitz Y, Goldberg O, Bello R, Cleper R, Lebenthal Y, Levy-Shraga Y, Tiosano D, Chezana A, Regev R, Zeitlin L. Nephrocalcinosis tendency does not worsen under burosumab treatment for X-linked hypophosphatemic rickets: a multicenter pediatric study. Front Pediatr 2024; 12:1487890. [PMID: 39687707 PMCID: PMC11646759 DOI: 10.3389/fped.2024.1487890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/30/2024] [Indexed: 12/18/2024] Open
Abstract
Background X-linked hypophosphatemic rickets (XLH) is associated with uninhibited FGF23 activity, which leads to phosphaturia, hypophosphatemia and depressed active vitamin D (1,25OH2D) levels. Conventional treatment with phosphate supplements and vitamin D analogs may lead to hypercalciuria (HC), nephrocalcinosis (NC) and hyperparathyroidism. We investigated the effects of burosumab treatment, an anti-FGF23 monoclonal antibody recently approved for XLH, on these complications. Methods This retrospective study included children with XLH who were treated with burosumab for at least one year at one of three referral centers. Clinical and biochemical potential treatment outcomes were regularly followed, including multiple urine calcium measurements and NC severity score (0 = no NC, 3 = worse NC). Results Twenty-six (13 male) children aged 7.6 ± 3.9 years were followed for 27.5 ± 9.6 months. Mean serum phosphate levels rapidly increased from 2.67 ± 0.61 at baseline to 3.57 ± 0.53 mg/dL after 3 months (p < 0.001) and remained stable thereafter. Concomitant decreases were observed in phosphaturia, serum alkaline phosphatase and parathyroid hormone. HC (U-Ca/Cr > 0.2 mg/mg) was detected in 2/26 (7.7%) patients before burosumab initiation, resolved in one and persisted, albeit improved, in the second. Two patients were newly diagnosed with HC, 15 and 3 months after therapy, which persisted in one of them despite dose reduction attempts. Seven patients had NC at baseline (mean score: 1.8 ± 0.34), but none showed deterioration or developed new NC. Conclusion In children with XLH treated with burosumab, HC was an infrequent side effect and preexisting NC did not worsen.
Collapse
Affiliation(s)
- Shelly Levi
- Pediatric Nephrology Institute, Schneider Children’s Medical Center, Petach Tikva, Israel
| | - Daniel Landau
- Pediatric Nephrology Institute, Schneider Children’s Medical Center, Petach Tikva, Israel
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Miriam Davidovits
- Pediatric Nephrology Institute, Schneider Children’s Medical Center, Petach Tikva, Israel
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Avivit Brener
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shoshana Gal
- Division of Pediatric Endocrinology, Ruth Rappaport Children’s Hospital, Rambam Medical Health Care Campus, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yael Borovitz
- Pediatric Nephrology Institute, Schneider Children’s Medical Center, Petach Tikva, Israel
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ori Goldberg
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Institute of Pulmonology, Schneider Children’s Medical Center, Petach Tikva, Israel
- Pediatric Pulmonary Service, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Rachel Bello
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children’s Medical Center, Petach Tikva, Israel
| | - Roxana Cleper
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Pediatric Nephrology Unit, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yael Lebenthal
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yael Levy-Shraga
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Pediatric Endocrinology Unit, The Edmond and Lily Safra Children’s Hospital, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Dov Tiosano
- Division of Pediatric Endocrinology, Ruth Rappaport Children’s Hospital, Rambam Medical Health Care Campus, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Adi Chezana
- Goldman School of Medicine, Faculty of Health Sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Ravit Regev
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Leonid Zeitlin
- The Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Pediatric Orthopedic Department, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
| |
Collapse
|
3
|
Rizzo M, Pennisi M, Macrì F, Falcone A, Di Pietro S, Mhalhel K, Giudice E. Bilateral Global Nephrocalcinosis in a Uremic Puppy. Vet Sci 2024; 11:338. [PMID: 39195792 PMCID: PMC11359828 DOI: 10.3390/vetsci11080338] [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: 06/06/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
This study explores kidney disease in young dogs, focusing on early diagnosis, management, and the importance of staging for effective treatment. Highlighting mineral metabolism imbalances and complications such as nephrocalcinosis, the study presents a case of severe renal failure with uremic syndrome and bilateral nephrocalcinosis in a 50-day-old puppy. Despite intensive care, the puppy's condition deteriorated rapidly, leading to euthanasia. The study underscores the challenges in diagnosing and managing canine nephrocalcinosis in young animals. It emphasizes the need for further research to improve the understanding and treatment outcomes in such cases, ultimately enhancing the quality of life for animals suffering from this rare condition.
Collapse
Affiliation(s)
- Maria Rizzo
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Melissa Pennisi
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Francesco Macrì
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Annastella Falcone
- Veterinary Teaching Hospital, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy;
| | - Simona Di Pietro
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Kamel Mhalhel
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Elisabetta Giudice
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| |
Collapse
|
4
|
Oh GJ, Butani L. Nephrocalcinosis in Neonates. Neoreviews 2024; 25:e88-e98. [PMID: 38296790 DOI: 10.1542/neo.25-2-e88] [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: 02/02/2024]
Abstract
Nephrocalcinosis occurs in as many as 40% of preterm neonates. Many causes and contributors predispose neonates to develop nephrocalcinosis, including metabolic, genetic, and iatrogenic factors. Because nephrocalcinosis can be a manifestation of an underlying genetic disorder, neonates with nephrocalcinosis must undergo an evaluation to identify and address contributors, to prevent further renal calcium deposition that can potentially lead to renal dysfunction. In this article, we review the epidemiology, pathogenesis, diagnosis, and evaluation of nephrocalcinosis in neonates. We also summarize the natural history of nephrocalcinosis of prematurity as well as the management of this condition.
Collapse
Affiliation(s)
- Gia J Oh
- Department of Pediatrics, Division of Pediatric Nephrology, University of California, Davis, Children's Hospital, Sacramento, CA
| | - Lavjay Butani
- Department of Pediatrics, Division of Pediatric Nephrology, University of California, Davis, Children's Hospital, Sacramento, CA
| |
Collapse
|
5
|
Eren E, Karabulut YY, Eren M, Kadir S. Mineralogy, geochemistry, and micromorphology of human kidney stones (urolithiasis) from Mersin, the southern Turkey. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023:10.1007/s10653-023-01525-8. [PMID: 36934357 DOI: 10.1007/s10653-023-01525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
This study describes the primary characteristics of the selected kidney stones surgically removed from the patients at the Mersin University Hospital in the southern Turkey and interprets their formation via petrographic, geochemical, XRD, SEM-EDX, and ICP-MS/OES analyses. The analytical results revealed that the kidney stones are composed of the minerals whewellite, struvite, hydroxyapatite, and uric acid alone or in different combinations. The samples occur in staghorn, bean-shaped composite, and individual rounded particle shapes, which are controlled by the shape of the nucleus and the site of stone formation. The cross-section of the samples shows concentric growth layers due to variations in saturation, characterizing the metastable phase. Kidney stone formation includes two main stages: (i) nucleation and (ii) aggregation and/or growth. Nucleation was either Randall plaque of hydroxyapatite in tissue on the surface of the papilla or a coating of whewellite on the plaque, or crystallization as free particles in the urine. Subsequently, aggregation or growth occurs by precipitation of stone-forming materials around the plaque or coating carried into the urine, or around the nucleus formed in situ in the urine. Urinary supersaturation is the main driving force of crystallization processes; and is controlled by many factors including bacterially induced supersaturation.
Collapse
Affiliation(s)
- Elif Eren
- Faculty of Medicine, San Raffaele University, Milan, Italy.
| | | | - Muhsin Eren
- Department of Geological Engineering, Mersin University, Mersin, Turkey
| | - Selahattin Kadir
- Department of Geological Engineering, Eskişehir Osmangazi University, Eskişehir, Turkey
| |
Collapse
|
6
|
Rocky road-an uncommon reason for kidney allograft dysfunction: Answers. Pediatr Nephrol 2021; 36:1461-1463. [PMID: 33025203 DOI: 10.1007/s00467-020-04688-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/07/2020] [Accepted: 06/18/2020] [Indexed: 10/23/2022]
|
7
|
Prajapati S, Tomar B, Srivastava A, Narkhede YB, Gaikwad AN, Lahiri A, Mulay SR. 6,7-Dihydroxycoumarin ameliorates crystal-induced necroptosis during crystal nephropathies by inhibiting MLKL phosphorylation. Life Sci 2021; 271:119193. [PMID: 33577856 DOI: 10.1016/j.lfs.2021.119193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 01/09/2023]
Abstract
AIMS Mineralization of crystalline particles and the formation of renal calculi contribute to the pathogenesis of crystal nephropathies. Several recent studies on the biology of crystal handling implicated intrarenal crystal deposition-induced necroinflammation in their pathogenesis. We hypothesized that 6,7-dihydroxycoumarin (DHC) inhibit intrarenal crystal cytotoxicity and necroinflammation, and ameliorate crystal-induced chronic kidney disease (CKD). MAIN METHODS An unbiased high content screening coupled with fluorescence microscopy was used to identify compounds that inhibit CaOx crystal cytotoxicity. The ligand-protein interactions were identified using computational models e.g. molecular docking and molecular dynamics simulations. Furthermore, mice and rat models of oxalate-induced CKD were used for in-vivo studies. Renal injury, crystal deposition, and fibrosis were assessed by histology analysis. Western blots were used to quantify the protein expression. Data were expressed as boxplots and analyzed using one way ANOVA. KEY FINDINGS An unbiased high-content screening in-vitro identified 6,7-DHC as a promising candidate. Further, 6,7-DHC protected human and mouse cells from calcium oxalate (CaOx) crystal-induced necroptosis in-vitro as well as mice and rats from oxalate-induced CKD in either preventive or therapeutic manner. Computational modeling demonstrated that 6,7-DHC interact with MLKL, the key protein in the necroptosis machinery, and inhibit its phosphorylation by ATP, which was evident in both in-vitro and in-vivo analyses. SIGNIFICANCE Together, our results indicate that 6,7-DHC possesses a novel pharmacological property as a MLKL inhibitor and could serve as a lead molecule for further development of coumarin-based novel MLKL inhibitors. Furthermore, our data identify 6,7-DHC as a novel therapeutic strategy to combat crystal nephropathies.
Collapse
Affiliation(s)
- Smita Prajapati
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Bhawna Tomar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anjali Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Yogesh B Narkhede
- Department of Bioengineering, Bourn's College of Engineering, University of California, CA 92521, USA
| | - Anil N Gaikwad
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amit Lahiri
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shrikant R Mulay
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
8
|
Kletzmayr A, Bigler M, Montanari E, Kuro-o M, Hayashi H, Ivarsson ME, Leroux JC. Development of a Kidney Calcification Inhibitor Employing Image-Based Profiling: A Proof-of-Concept Study. ACS Pharmacol Transl Sci 2020; 3:1339-1351. [DOI: 10.1021/acsptsci.0c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Anna Kletzmayr
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Melina Bigler
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Elita Montanari
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Makoto Kuro-o
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Hirosaka Hayashi
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | | | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
9
|
Klinkhammer BM, Djudjaj S, Kunter U, Palsson R, Edvardsson VO, Wiech T, Thorsteinsdottir M, Hardarson S, Foresto-Neto O, Mulay SR, Moeller MJ, Jahnen-Dechent W, Floege J, Anders HJ, Boor P. Cellular and Molecular Mechanisms of Kidney Injury in 2,8-Dihydroxyadenine Nephropathy. J Am Soc Nephrol 2020; 31:799-816. [PMID: 32086278 DOI: 10.1681/asn.2019080827] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hereditary deficiency of adenine phosphoribosyltransferase causes 2,8-dihydroxyadenine (2,8-DHA) nephropathy, a rare condition characterized by formation of 2,8-DHA crystals within renal tubules. Clinical relevance of rodent models of 2,8-DHA crystal nephropathy induced by excessive adenine intake is unknown. METHODS Using animal models and patient kidney biopsies, we assessed the pathogenic sequelae of 2,8-DHA crystal-induced kidney damage. We also used knockout mice to investigate the role of TNF receptors 1 and 2 (TNFR1 and TNFR2), CD44, or alpha2-HS glycoprotein (AHSG), all of which are involved in the pathogenesis of other types of crystal-induced nephropathies. RESULTS Adenine-enriched diet in mice induced 2,8-DHA nephropathy, leading to progressive kidney disease, characterized by crystal deposits, tubular injury, inflammation, and fibrosis. Kidney injury depended on crystal size. The smallest crystals were endocytosed by tubular epithelial cells. Crystals of variable size were excreted in urine. Large crystals obstructed whole tubules. Medium-sized crystals induced a particular reparative process that we term extratubulation. In this process, tubular cells, in coordination with macrophages, overgrew and translocated crystals into the interstitium, restoring the tubular luminal patency; this was followed by degradation of interstitial crystals by granulomatous inflammation. Patients with adenine phosphoribosyltransferase deficiency showed similar histopathological findings regarding crystal morphology, crystal clearance, and renal injury. In mice, deletion of Tnfr1 significantly reduced tubular CD44 and annexin two expression, as well as inflammation, thereby ameliorating the disease course. In contrast, genetic deletion of Tnfr2, Cd44, or Ahsg had no effect on the manifestations of 2,8-DHA nephropathy. CONCLUSIONS Rodent models of the cellular and molecular mechanisms of 2,8-DHA nephropathy and crystal clearance have clinical relevance and offer insight into potential future targets for therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Thorsten Wiech
- Institute of Pathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; and
| | | | - Sverrir Hardarson
- Department of Pathology Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Shrikant R Mulay
- Division of Nephrology, Klinikum der Universität, LMU München, Munich, Germany
| | | | | | | | - Hans-Joachim Anders
- Division of Nephrology, Klinikum der Universität, LMU München, Munich, Germany
| | - Peter Boor
- Institute of Pathology, .,Division of Nephrology and Immunology.,Department of Electron Microscopy, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
10
|
Zhang Y, Zhu R, Liu D, Gong M, Hu W, Yi Q, Zhang J. Tetracycline attenuates calcifying nanoparticles-induced renal epithelial injury through suppression of inflammation, oxidative stress, and apoptosis in rat models. Transl Androl Urol 2019; 8:619-630. [PMID: 32038958 DOI: 10.21037/tau.2019.11.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Calcifying nanoparticles (CNPs) has been associated with the occurrence and development of kidney stones, but the exact mechanism is not clear. This study aimed to establish a rat model of CNP-induced renal epithelial injury and assess the efficacy of tetracycline in preventing this injury. Methods Kidney stones from patients after percutaneous nephrolithotomy (PCNL) were collected to isolate and culture CNPs. Thirty Sprague-Dawley rats were divided into three groups: the sham group (G1), the CNP group (G2), and the CNP + tetracycline group (G3). Rats in G2 and G3 were given an intravenous injection of CNPs via the tail vein, while rats in G1 were given saline. Meanwhile, rats in G3 were given tetracycline by gavage twice a day at a dose of 25 mg/kg. After 8 weeks, the 24-h urine of all rats was collected, and all rats were sacrificed to obtain blood and kidneys. Results The results revealed that in G2, activities of antioxidant enzymes such as superoxide dismutase and catalase were significantly lower than those in G1, while malondialdehyde activity in G2 was significantly higher than that in G1 and both of them were inhibited by tetracycline co-treatment in G3. CNPs significantly increased expression of inflammatory cytokines, including monocyte chemotactic protein 1 and interleukin 6, which were largely alleviated in G3. CNPs significantly increased TUNEL-positive cells and the apoptosis activity of Bcl2-associated X protein but decreased B-cell lymphoma-2 level compared with that in G1, and was limited by tetracycline co-treatment in G3. Furthermore, CNPs led to notable renal tubular epithelial cell damage, hyaline cast formation, desquamation, swelling, vacuolization in histology, all of which were alleviated by tetracycline. Conclusions Tetracycline can attenuate CNP-induced renal epithelial injury through suppression of inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Yuqing Zhang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Dong Liu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Min Gong
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Wei Hu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Qingtong Yi
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Jie Zhang
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| |
Collapse
|
11
|
Torres JA, Rezaei M, Broderick C, Lin L, Wang X, Hoppe B, Cowley BD, Savica V, Torres VE, Khan S, Holmes RP, Mrug M, Weimbs T. Crystal deposition triggers tubule dilation that accelerates cystogenesis in polycystic kidney disease. J Clin Invest 2019; 129:4506-4522. [PMID: 31361604 PMCID: PMC6763267 DOI: 10.1172/jci128503] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
The rate of disease progression in autosomal-dominant (AD) polycystic kidney disease (PKD) exhibits high intra-familial variability suggesting that environmental factors may play a role. We hypothesized that a prevalent form of renal insult may accelerate cystic progression and investigated tubular crystal deposition. We report that calcium oxalate (CaOx) crystal deposition led to rapid tubule dilation, activation of PKD-associated signaling pathways, and hypertrophy in tubule segments along the affected nephrons. Blocking mTOR signaling blunted this response and inhibited efficient excretion of lodged crystals. This mechanism of "flushing out" crystals by purposefully dilating renal tubules has not previously been recognized. Challenging PKD rat models with CaOx crystal deposition, or inducing calcium phosphate deposition by increasing dietary phosphorous intake, led to increased cystogenesis and disease progression. In a cohort of ADPKD patients, lower levels of urinary excretion of citrate, an endogenous inhibitor of calcium crystal formation, correlated with increased disease severity. These results suggest that PKD progression may be accelerated by commonly occurring renal crystal deposition which could be therapeutically controlled by relatively simple measures.
Collapse
Affiliation(s)
- Jacob A. Torres
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Mina Rezaei
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Caroline Broderick
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Louis Lin
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Xiaofang Wang
- Mayo Clinic College of Medicine, Division of Nephrology and Hypertension, Rochester, Minnesota, USA
| | - Bernd Hoppe
- University Children’s Hospital Bonn, Division of Pediatric Nephrology, Bonn, Germany
| | - Benjamin D. Cowley
- University of Oklahoma Health Sciences Center, Department of Medicine, Section of Nephrology, Oklahoma City, Oklahoma, USA
| | - Vincenzo Savica
- University of Messina, Department of Clinical and Experimental Medicine, Messina, Italy
| | - Vicente E. Torres
- Mayo Clinic College of Medicine, Division of Nephrology and Hypertension, Rochester, Minnesota, USA
| | - Saeed Khan
- University of Florida, Department of Pathology, Gainesville, Florida, USA
| | | | - Michal Mrug
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | - Thomas Weimbs
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| |
Collapse
|
12
|
Gorvin CM, Ahmad BN, Stechman MJ, Loh NY, Hough TA, Leo P, Marshall M, Sethi S, Bentley L, Piret SE, Reed A, Jeyabalan J, Christie PT, Wells S, Simon MM, Mallon AM, Schulz H, Huebner N, Brown MA, Cox RD, Brown SD, Thakker RV. An N-Ethyl-N-Nitrosourea (ENU)-Induced Tyr265Stop Mutation of the DNA Polymerase Accessory Subunit Gamma 2 (Polg2) Is Associated With Renal Calcification in Mice. J Bone Miner Res 2019; 34:497-507. [PMID: 30395686 PMCID: PMC6446808 DOI: 10.1002/jbmr.3624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/12/2018] [Accepted: 10/28/2018] [Indexed: 12/24/2022]
Abstract
Renal calcification (RCALC) resulting in nephrolithiasis and nephrocalcinosis, which affects ∼10% of adults by 70 years of age, involves environmental and genetic etiologies. Thus, nephrolithiasis and nephrocalcinosis occurs as an inherited disorder in ∼65% of patients, and may be associated with endocrine and metabolic disorders including: primary hyperparathyroidism, hypercalciuria, renal tubular acidosis, cystinuria, and hyperoxaluria. Investigations of families with nephrolithiasis and nephrocalcinosis have identified some causative genes, but further progress is limited as large families are unavailable for genetic studies. We therefore embarked on establishing mouse models for hereditary nephrolithiasis and nephrocalcinosis by performing abdominal X-rays to identify renal opacities in N-ethyl-N-nitrosourea (ENU)-mutagenized mice. This identified a mouse with RCALC inherited as an autosomal dominant trait, designated RCALC type 2 (RCALC2). Genomewide mapping located the Rcalc2 locus to a ∼16-Mbp region on chromosome 11D-E2 and whole-exome sequence analysis identified a heterozygous mutation in the DNA polymerase gamma-2, accessory subunit (Polg2) resulting in a nonsense mutation, Tyr265Stop (Y265X), which co-segregated with RCALC2. Kidneys of mutant mice (Polg2+/Y265X ) had lower POLG2 mRNA and protein expression, compared to wild-type littermates (Polg2+/+ ). The Polg2+/Y265X and Polg2+/+ mice had similar plasma concentrations of sodium, potassium, calcium, phosphate, chloride, urea, creatinine, glucose, and alkaline phosphatase activity; and similar urinary fractional excretion of calcium, phosphate, oxalate, and protein. Polg2 encodes the minor subunit of the mitochondrial DNA (mtDNA) polymerase and the mtDNA content in Polg2+/Y265X kidneys was reduced compared to Polg2+/+ mice, and cDNA expression profiling revealed differential expression of 26 genes involved in several biological processes including mitochondrial DNA function, apoptosis, and ubiquitination, the complement pathway, and inflammatory pathways. In addition, plasma of Polg2+/Y265X mice, compared to Polg2+/+ littermates had higher levels of reactive oxygen species. Thus, our studies have identified a mutant mouse model for inherited renal calcification associated with a Polg2 nonsense mutation. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Bushra N Ahmad
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michael J Stechman
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Nellie Y Loh
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Tertius A Hough
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Paul Leo
- Translational Genomics Group, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology at Translational Research Institute, Brisbane, Australia
| | - Mhairi Marshall
- Translational Genomics Group, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology at Translational Research Institute, Brisbane, Australia
| | - Siddharth Sethi
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Liz Bentley
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Sian E Piret
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Anita Reed
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jeshmi Jeyabalan
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul T Christie
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sara Wells
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Michelle M Simon
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Ann-Marie Mallon
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Herbert Schulz
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Matthew A Brown
- Translational Genomics Group, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology at Translational Research Institute, Brisbane, Australia
| | - Roger D Cox
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Steve D Brown
- Mary Lyon Centre and Mammalian Genetics Unit, Medical Research Council, Harwell, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Priante G, Ceol M, Gianesello L, Furlan C, Del Prete D, Anglani F. Human proximal tubular cells can form calcium phosphate deposits in osteogenic culture: role of cell death and osteoblast-like transdifferentiation. Cell Death Discov 2019; 5:57. [PMID: 30701089 PMCID: PMC6349935 DOI: 10.1038/s41420-019-0138-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
Nephrocalcinosis is a clinicopathological entity characterized by microscopic calcium crystals in the renal parenchyma, within the tubular lumen or in the interstitium. Crystal binding to tubular cells may be the cause underlying nephrocalcinosis and nephrolithiasis. Pathological circumstances, such as acute cortical necrosis, may induce healthy cells to acquire a crystal-binding phenotype. The present study aimed to investigate whether human renal proximal tubular cells (HK-2 cells) can form calcium phosphate deposits under osteogenic conditions, and whether apoptosis and/or osteogenic-like processes are involved in cell calcification. HK-2 cells were cultured in standard or osteogenic medium for 1, 5, and 15 days. Von Kossa staining and ESEM were used to analyze crystal deposition. Apoptosis was investigated, analyzing caspase activation by in-cell Western assay, membrane translocation of phosphotidylserine by annexin V-FITC/propidium iodide staining, and DNA fragmentation by TUNEL assay. qRT/PCR, immunolabeling and cytochemistry were performed to assess osteogenic activation (Runx2, Osteonectin, Osteopontin and ALP), and early genes of apoptosis (BAX, Bcl-2). HK-2 cell mineralization was successfully induced on adding osteogenic medium. Calcium phosphate deposition increased in a time-dependent manner, and calcified cell aggregates exhibited characteristic signs of apoptosis. At 15 days, calcifying HK-2 cells revealed osteogenic markers, such as Runx2, ALP, osteonectin and osteopontin. Monitoring the processes at 1, 5, and 15 days showed apoptosis starting already after 5 days of osteogenic induction, when the first small calcium phosphate crystals began to appear on areas where cell aggregates were in apoptotic conditions. The cell death process proved caspase-dependent. The importance of apoptosis was reinforced by the time-dependent increase in BAX expression, starting from day 1. These findings strongly support the hypothesis that apoptosis triggered HK-2 calcification even before any calcium phosphate crystal deposition or acquisition of an osteogenic phenotype.
Collapse
Affiliation(s)
- Giovanna Priante
- Laboratory of Kidney Histomorphology and Molecular Biology, Clinical Nephrology, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Monica Ceol
- Laboratory of Kidney Histomorphology and Molecular Biology, Clinical Nephrology, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Lisa Gianesello
- Laboratory of Kidney Histomorphology and Molecular Biology, Clinical Nephrology, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Claudio Furlan
- Center for ESEM and SEM analyses (CEASC), University of Padova, Padova, Italy
| | - Dorella Del Prete
- Laboratory of Kidney Histomorphology and Molecular Biology, Clinical Nephrology, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Franca Anglani
- Laboratory of Kidney Histomorphology and Molecular Biology, Clinical Nephrology, Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
14
|
Meeting report of the "Symposium on kidney stones and mineral metabolism: calcium kidney stones in 2017". J Nephrol 2019; 32:681-698. [PMID: 30680550 DOI: 10.1007/s40620-019-00587-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
A symposium on kidney stones and mineral metabolism held on December 2017 in Brussels, Belgium was the first international multidisciplinary conference of the International Collaborative Network on Kidney Stones and Mineral Metabolism. This meeting addressed epidemiology, underlying pathophysiological mechanisms, genetics, pathological, as well as clinical and research topics. The participants included clinicians and recognized experts in the field from Europe and the United States interacted closely during the symposium which promoted a chance to explore new frontiers in the field of kidney stone disease. This manuscript summarizes some of the major highlights of the meeting.
Collapse
|
15
|
Abstract
Calcium kidney stones are common worldwide. Most are idiopathic and composed of calcium oxalate. Calcium phosphate is present in around 80% and may initiate stone formation. Stone production is multifactorial with a polygenic genetic contribution. Phosphaturia is found frequently among stone formers but until recently received scant attention. This review examines possible mechanisms for the phosphaturia and its relevance to stone formation from a wide angle. There is a striking lack of clinical data. Phosphaturia is associated, but not correlated, with hypercalciuria, increased 1,25 dihydroxy-vitamin D [1,25 (OH)2D], and sometimes evidence of disturbances in proximal renal tubular function. Phosphate reabsorption in the proximal renal tubules requires tightly regulated interaction of many proteins. Paracellular flow through intercellular tight junctions is the major route of phosphate absorption from the intestine and can be reduced therapeutically in hyperphosphatemic patients. In monogenic defects stones develop when phosphaturia is associated with hypercalciuria, generally explained by increased 1,25 (OH)2D production in response to hypophosphatemia. Calcification does not occur in disorders with increased FGF23 when phosphaturia occurs in isolation and 1,25 (OH)2D is suppressed. Candidate gene studies have identified mutations in the phosphate transporters, but in few individuals. One genome-wide study identified a polymorphism of the phosphate transporter gene SLC34A4 associated with stones. Others did not find mutations obviously linked to phosphate reabsorption. Future genetic studies should have a wide trawl and should focus initially on groups of patients with clearly defined phenotypes. The global data should be pooled.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.
| |
Collapse
|
16
|
Khamchun S, Thongboonkerd V. Cell cycle shift from G0/G1 to S and G2/M phases is responsible for increased adhesion of calcium oxalate crystals on repairing renal tubular cells at injured site. Cell Death Discov 2018; 4:106. [PMID: 30774989 PMCID: PMC6374384 DOI: 10.1038/s41420-018-0123-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/24/2018] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
Renal tubular cell injury can enhance calcium oxalate monohydrate (COM) crystal adhesion at the injured site and thus may increase the stone risk. Nevertheless, underlying mechanism of such enhancement remained unclear. In the present study, confluent MDCK renal tubular cell monolayers were scratched to allow cells to proliferate and repair the injured site. At 12-h post-scratch, the repairing cells had significant increases in crystal adhesion capacity and cell proliferation as compared to the control. Cell cycle analysis using flow cytometry demonstrated that the repairing cells underwent cell cycle shift from G0/G1 to S and G2/M phases. Cyclosporin A (CsA) and hydroxyurea (HU) at sub-toxic doses caused cell cycle shift mimicking that observed in the repairing cells. Crystal-cell adhesion assay confirmed the increased crystal adhesion capacity of the CsA-treated and HU-treated cells similar to that of the repairing cells. These findings provide evidence indicating that cell cycle shift from G0/G1 to S and G2/M phases is responsible, at least in part, for the increased adhesion of COM crystals on repairing renal tubular cells at the injured site.
Collapse
Affiliation(s)
- Supaporn Khamchun
- 1Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,2Department of Immunology and Immunology Graduate Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- 1Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,3Center for Research in Complex Systems Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
17
|
Mulay SR, Shi C, Ma X, Anders HJ. Novel Insights into Crystal-Induced Kidney Injury. KIDNEY DISEASES 2018; 4:49-57. [PMID: 29998119 DOI: 10.1159/000487671] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/14/2018] [Indexed: 01/04/2023]
Abstract
Background The entity of crystal nephropathies encompasses a spectrum of different kidney injuries induced by crystal-formed intrinsic minerals, metabolites, and proteins or extrinsic dietary components and drug metabolites. Depending on the localization and dynamics of crystal deposition, the clinical presentation can be acute kidney injury, progressive chronic kidney disease, or renal colic. Summary The molecular mechanisms involving crystal-induced injury are diverse and remain poorly understood. Type 1 crystal nephropathies arise from crystals in the vascular lumen (cholesterol embolism) or the vascular wall (atherosclerosis) and involve kidney infarcts or chronic ischemia, respectively. Type 2 crystal nephropathies arise from intratubular crystal deposition causing obstruction, interstitial inflammation, and tubular cell injury. NLRP3 inflammasome and necroptosis drive renal necroinflammation in acute settings. Type 3 is represented by crystal and stone formation in the draining urinary tract, i.e., urolithiasis, causing renal colic and chronic obstruction. Key Messages Dissecting the types of injury is the first step towards a better understanding of the pathophysiology of crystal nephropathies. Crystal-induced acti-vation of the inflammasome and necroptosis, crystal adhesion, crystallization inhibitors, extratubulation, and granulo-ma formation are only a few of certainly many involved pathomechanisms that deserve further studies to eventually form the basis for innovative cures for these diseases.
Collapse
Affiliation(s)
- Shrikant R Mulay
- Division of Nephrology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Chongxu Shi
- Division of Nephrology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Xiaoyuan Ma
- Division of Nephrology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Hans Joachim Anders
- Division of Nephrology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| |
Collapse
|
18
|
Priante G, Quaggio F, Gianesello L, Ceol M, Cristofaro R, Terrin L, Furlan C, Del Prete D, Anglani F. Caspase-independent programmed cell death triggers Ca 2PO 4 deposition in an in vitro model of nephrocalcinosis. Biosci Rep 2018; 38:BSR20171228. [PMID: 29208768 PMCID: PMC5770611 DOI: 10.1042/bsr20171228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 01/12/2023] Open
Abstract
Nephrocalcinosis involves the deposition of microscopic crystals in the tubular lumen or interstitium. While the clinical, biochemical, and genetic aspects of the diseases causing nephrocalcinosis have been elucidated, little is known about the cellular events in this calcification process. We previously reported a phenomenon involving the spontaneous formation of Ca2PO4 nodules in primary papillary renal cells from a patient with medullary nephrocalcinosis harboring a rare glial cell-derived neurotrophic factor (GDNF) gene variant. We also demonstrated that cultivating GDNF-silenced human kidney-2 (HK-2) cells in osteogenic conditions for 15 days triggered Ca2PO4 deposits. Given the reportedly close relationship between cell death and pathological calcification, aim of the present study was to investigate whether apoptosis is involved in the calcification of GDNF-silenced HK-2 cells under osteogenic conditions. Silenced and control cells were cultured in standard and osteogenic medium for 1, 5, and 15 days, and any Ca2PO4 deposition was identified by means of von Kossa staining and environmental SEM (ESEM) analyses. Based on the results of annexin V and propidium iodide (PI) analysis, and terminal deoxynucleotidyl transferase dUTP-biotin nick end labeling (TUNEL) assay, the silenced cells in the osteogenic medium showed a significant increase in the percentage of cells in the late phase of apoptosis and an increased Ca2PO4 deposition at 15 days. The results of quantitative real-time PCR (qRT-PCR) of BAX and BCL2, and in-cell Western analysis of caspases indicated that the cell death process was independent of caspase-3, -6, -7, and -9 activation, however. Using this model, we provide evidence of caspase-independent cell death triggering the calcification process in GDNF-silenced HK-2 cells.
Collapse
Affiliation(s)
- Giovanna Priante
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Federica Quaggio
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Lisa Gianesello
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Monica Ceol
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Rosalba Cristofaro
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Liliana Terrin
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Claudio Furlan
- Center for Laboratory Analyses and Certification Services (CEASC), University of Padua, Padua, Italy
| | - Dorella Del Prete
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| | - Franca Anglani
- Department of Medicine - DIMED, Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, University of Padua, Padua, Italy
| |
Collapse
|
19
|
Albert A, Tiwari V, Paul E, Ponnusamy S, Ganesan D, Prabhakaran R, Mariaraj Sivakumar S, Govindan Sadasivam S. Oral administration of oxalate-enriched spinach extract as an improved methodology for the induction of dietary hyperoxaluric nephrocalcinosis in experimental rats. Toxicol Mech Methods 2017; 28:195-204. [DOI: 10.1080/15376516.2017.1388459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Abhishek Albert
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Vidhi Tiwari
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Eldho Paul
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Sasikumar Ponnusamy
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Divya Ganesan
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Rajkumar Prabhakaran
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Selvi Mariaraj Sivakumar
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Selvam Govindan Sadasivam
- Department of Biochemistry, Centre for Excellence in Genomics Science, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
20
|
Cheunsuchon B, Sritippayawan S. Successful treatment of early allograft dysfunction with cinacalcet in a patient with nephrocalcinosis caused by severe hyperparathyroidism: a case report. BMC Res Notes 2017; 10:153. [PMID: 28390426 PMCID: PMC5385015 DOI: 10.1186/s13104-017-2477-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 04/04/2017] [Indexed: 01/05/2023] Open
Abstract
Background Hyperparathyroidism is common in patients undergoing kidney transplantation. Occasionally, this condition can cause early allograft dysfunction by inducing calcium phosphate deposition in the allograft, which results in nephrocalcinosis. Although nephrocalcinosis occurs occasionally in kidney allografts, it has only rarely been reported in the literature. Case presentation Here, we present the case of a 58-year-old Thai woman with severe hyperparathyroidism who received a living-related kidney transplant from her 35-year-old son. Our patient developed allograft dysfunction on day 2 post-transplantation despite good functioning graft on day 1. Allograft biopsy showed extensive calcium phosphate deposition in distal tubules. She was treated with cinacalcet (a calcimimetic agent) and aluminum hydroxide. Allograft function was restored to normal within 1 week after transplantation with greatly reduced intact parathyroid hormone level. Conclusion Hyperparathyroidism in early functioning allograft causes elevated calcium and phosphate concentration in distal tubules resulting in nephrocalcinosis. The massive calcium phosphate precipitation obstructs tubular lumens, which leads to acute tubular dysfunction. Treatment of nephrocalcinosis with cinacalcet is safe and may improve this condition by increasing serum phosphate and reducing serum calcium and intact parathyroid hormone.
Collapse
Affiliation(s)
- Boonyarit Cheunsuchon
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkok, 10700, Thailand.
| | - Suchai Sritippayawan
- Division of Nephrology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
21
|
Haymann JP, Vinsonneau C, Girshovich A, Daudon M. Insuffisance rénale aiguë obstructive : une lecture physiopathologique. Nephrol Ther 2017; 13 Suppl 1:S1-S5. [DOI: 10.1016/j.nephro.2017.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/08/2017] [Indexed: 11/27/2022]
|
22
|
Mulay SR, Anders HJ. Crystal nephropathies: mechanisms of crystal-induced kidney injury. Nat Rev Nephrol 2017; 13:226-240. [DOI: 10.1038/nrneph.2017.10] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Renal dysfunction in primary hyperparathyroidism; effect of Parathyroidectomy: A retrospective Cohort Study. Int J Surg 2016; 36:383-387. [DOI: 10.1016/j.ijsu.2016.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/02/2016] [Indexed: 01/16/2023]
|
24
|
Martel J, Wu CY, Young JD. Translocation of mineralo-organic nanoparticles from blood to urine: a new mechanism for the formation of kidney stones? Nanomedicine (Lond) 2016; 11:2399-404. [DOI: 10.2217/nnm-2016-0246] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent studies indicate that mineralo-organic nanoparticles form in various human body fluids, including blood and urine. These nanoparticles may form within renal tubules and increase in size in supersaturated urine, eventually leading to the formation of kidney stones. Here, we present observations suggesting that mineralo-organic nanoparticles found in blood may induce kidney stone formation via an alternative mechanism in which the particles translocate through endothelial and renal epithelial cells to reach urine. We propose that this alternative mechanism of kidney stone formation and the study of mineralo-organic nanoparticles in general may provide novel strategies for the early detection and treatment of ectopic calcifications and kidney stones.
Collapse
Affiliation(s)
- Jan Martel
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan 33302, Taiwan
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan 33302, Taiwan
| | - Cheng-Yeu Wu
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan 33302, Taiwan
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan 33302, Taiwan
| | - John D Young
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan 33302, Taiwan
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan 33302, Taiwan
- Laboratory of Cellular Physiology & Immunology, Rockefeller University, New York, NY 10021, USA
- Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| |
Collapse
|
25
|
Cao Y, Liu W, Hui L, Zhao J, Yang X, Wang Y, Niu H. Renal tubular injury induced by ischemia promotes the formation of calcium oxalate crystals in rats with hyperoxaluria. Urolithiasis 2016; 44:389-97. [PMID: 27040948 DOI: 10.1007/s00240-016-0876-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 03/22/2016] [Indexed: 11/25/2022]
Abstract
Hyperoxaluria and cell injury are key factors in urolithiasis. Oxalate metabolism may be altered by renal dysfunction and therefore, impact the deposition of calcium oxalate (CaOx) crystals. We investigated the relationship of renal function, oxalate metabolism and CaOx crystal deposition in renal ischemia. One hundred male Sprague-Dawley rats were randomly divided into four groups. Hyperoxaluria model (Group A and B) was established by feeding rats with 0.75 % ethylene glycol (EG). The left renal pedicle was clamped for 30 min to establish renal ischemia Groups (B and C), while Groups A and D underwent sham operation. Then, serum and urine oxalate (Ox), creatinine (Cr) and urea nitrogen (UN) levels were evaluated by liquid chromatography mass spectrometry (LCMS) and ion mass spectrum (IMS) at days 0, 2, 4, 7, and 14. CaOx crystallization was assessed by transmission electron microscope (TEM). A temporal and significant increase of serum Cr and UN levels was observed in Groups B and C compared to values obtained for Groups A and D (P < 0.05). Ox levels in serum and urine were significantly higher in Groups A and B than in the other two groups from day 7 (P < 0.05). In addition, CaOx crystallization was observed in both Groups A and B, but Group B showed earlier and more pronounced crystal deposition in the renal tissue. Our results indicated that renal tubular injury induced by renal ischemia might not affect Ox levels but could promote CaOx crystal retention under hyperoxaluria.
Collapse
Affiliation(s)
- Yanwei Cao
- Department of Urology, The Affiliated Hospital of Qingdao University, Haier Road No. 59, Qingdao, 266101, Shandong Province, People's Republic of China
| | - Wanpeng Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Limei Hui
- Department of Urology, Affiliated Hospital of Hebei University of Engineering, Congtai Road No. 81, Handan, 056002, Hebei Province, People's Republic of China
| | - Jianjun Zhao
- Department of Urology, Affiliated Hospital of Hebei University of Engineering, Congtai Road No. 81, Handan, 056002, Hebei Province, People's Republic of China
| | - Xuecheng Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Haier Road No. 59, Qingdao, 266101, Shandong Province, People's Republic of China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Haier Road No. 59, Qingdao, 266101, Shandong Province, People's Republic of China
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Haier Road No. 59, Qingdao, 266101, Shandong Province, People's Republic of China.
| |
Collapse
|
26
|
Piccoli GB, De Pascale A, Randone O, Vigotti FN, Priola AM, Naretto C, Ferraresi M, Aroasio E, Gonella S, Mongilardi E, Scognamiglio S, Consiglio V, Roggero S, Piga A, Roccatello D, Veltri A. Revisiting nephrocalcinosis: A single-centre perspective. A northern Italian experience. Nephrology (Carlton) 2016; 21:97-107. [DOI: 10.1111/nep.12535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Giorgina Barbara Piccoli
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Agostino De Pascale
- Radiology; ASOU San Luigi; Department of Oncology; University of Torino; Torino Italy
| | - Olga Randone
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Federica Neve Vigotti
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | | | - Carla Naretto
- Interregional Center for Rare Diseases; G Bosco Hospital; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Martina Ferraresi
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Emiliano Aroasio
- Laboratory; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Silvana Gonella
- Laboratory; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Elena Mongilardi
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Stefania Scognamiglio
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Valentina Consiglio
- SS Nephrology; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Simona Roggero
- Microcythemia Unit; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Antonio Piga
- Microcythemia Unit; ASOU San Luigi; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Dario Roccatello
- Interregional Center for Rare Diseases; G Bosco Hospital; Department of Clinical and Biological Sciences; University of Torino; Torino Italy
| | - Andrea Veltri
- Radiology; ASOU San Luigi; Department of Oncology; University of Torino; Torino Italy
| |
Collapse
|
27
|
Yavuz S, Bayazıt AK, Anarat A, Gonlusen G, Alsancak P. Medullary nephrocalcinosis in Schimke immuno-osseous dysplasia. Pediatr Int 2015; 57:310-3. [PMID: 25868949 DOI: 10.1111/ped.12455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/26/2014] [Accepted: 07/11/2014] [Indexed: 12/16/2022]
Abstract
Schimke immuno-osseous dysplasia (SIOD) is a rare hereditary disease characterized by skeletal dysplasia, immune deficiency and progressive renal disease. Kidney involvement mainly determines the prognosis. The most common renal pathology is focal segmental glomerulosclerosis (FSGS). Medullary nephrocalcinosis refers to the diffuse deposition of calcium salts in renal medulla and has not previously been identified in SIOD. Here we report the first case of a pediatric patient having typical features of SIOD with medullary nephrocalcinosis.
Collapse
Affiliation(s)
- Sevgi Yavuz
- Division of Pediatric Nephrology, Cukurova University Hospital, Adana, Turkey
| | | | | | | | | |
Collapse
|
28
|
Mezzabotta F, Cristofaro R, Ceol M, Del Prete D, Priante G, Familiari A, Fabris A, D'Angelo A, Gambaro G, Anglani F. Spontaneous calcification process in primary renal cells from a medullary sponge kidney patient harbouring a GDNF mutation. J Cell Mol Med 2015; 19:889-902. [PMID: 25692823 PMCID: PMC4395202 DOI: 10.1111/jcmm.12514] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 11/18/2014] [Indexed: 11/30/2022] Open
Abstract
Medullary nephrocalcinosis is a hallmark of medullary sponge kidney (MSK). We had the opportunity to study a spontaneous calcification process in vitro by utilizing the renal cells of a patient with MSK who was heterozygous for the c.-27 + 18G>A variant in the GDNF gene encoding glial cell-derived neurotrophic factor. The cells were obtained by collagenase digestion of papillary tissues from the MSK patient and from two patients who had no MSK or nephrocalcinosis. These cells were typed by immunocytochemistry, and the presence of mineral deposits was studied using von Kossa staining, scanning electron microscopy analysis and an ALP assay. Osteoblastic lineage markers were studied using immunocytochemistry and RT-PCR. Staminality markers were also analysed using flow cytometry, magnetic cell separation technology, immunocytochemistry and RT-PCR. Starting from p2, MSK and control cells formed nodules with a behaviour similar to that of calcifying pericytes; however, Ca2PO4 was only found in the MSK cultures. The MSK cells had morphologies and immunophenotypes resembling those of pericytes or stromal stem cells and were positive for vimentin, ZO1, αSMA and CD146. In addition, the MSK cells expressed osteocalcin and osteonectin, indicating an osteoblast-like phenotype. In contrast to the control cells, GDNF was down-regulated in the MSK cells. Stable GDNF knockdown was established in the HK2 cell line and was found to promote Ca2PO4 deposition when the cells were incubated with calcifying medium by regulating the osteonectin/osteopontin ratio in favour of osteonectin. Our data indicate that the human papilla may be a perivascular niche in which pericyte/stromal-like cells can undergo osteogenic differentiation under particular conditions and suggest that GDNF down-regulation may have influenced the observed phenomenon.
Collapse
Affiliation(s)
- Federica Mezzabotta
- Laboratory of Histomorphology and Molecular Biology of the Kidney, Nephrology Division, Department of Medicine DIMED, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Screening of different probiotic strains for their in vitro ability to metabolise oxalates: any prospective use in humans? J Clin Gastroenterol 2014; 48 Suppl 1:S91-5. [PMID: 25291139 DOI: 10.1097/mcg.0000000000000228] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Oxalate is the salt-forming ion of oxalic acid and can generate oxalate salts combining with various cations, such as sodium, potassium, magnesium, and calcium. Approximately 75% of all kidney stones are composed primarily of calcium oxalate (CaOx) and hyperoxaluria, a condition involving high urinary oxalate concentration, is considered a primary risk factor for kidney stone formation, known as nephrolithiasis. Current therapeutic strategies often fail in their compliance or effectiveness, and CaOx stone recurrence is still common. After an initial stone, there is a 50% chance of forming a second stone within 7 years if the condition is left untreated. The potential therapeutic application of some probiotics, mainly lactobacilli and bifidobacteria, in reducing hyperoxaluria in vivo through intestinal oxalate degrading activity is compelling and initial reports are promising. This study was undertaken to screen different Lactobacillus and Bifidobacterium strains for their capacity to degrade oxalate in vitro using reverse-phase high-performance liquid chromatography (HPLC). METHODS The oxalate-degrading activity of 13 lactobacilli and 5 bifidobacteria was tested using a novel HPLC method after growth in a broth culture added with 10 mM ammonium oxalate. Experiments were repeated 3 times. Oxalobacter formigenes (DSM 4420) was used as positive reference to validate HPLC oxalate-degrading capability assays. RESULTS Lactobacillus strains were more efficient than bifidobacteria in degrading oxalates. L. paracasei LPC09 (DSM 24243) gave the best result, as 68.5% of ammonium oxalate was converted at the end of incubation, whereas the following best converters belong to the L. gasseri and L. acidophilus species. The relatively low conversion rate observed for most bifidobacteria can probably be attributed to intrinsic oxalate toxicity toward this genus. CONCLUSIONS Humans lack the enzymes needed to directly metabolise oxalate, and this potentially toxic compound is, therefore, managed using alternative pathways. As oxalate-degrading bacteria are present in the endogenous microbiota of the human intestine, although with significant individual differences, it is possible to hypothesise that the administration of selected oxalate-degrading probiotics could be an alternative and innovative approach to reducing the intestinal absorption of oxalate and the resulting urinary excretion.
Collapse
|
30
|
Biomimetic Randall's plaque as an in vitro model system for studying the role of acidic biopolymers in idiopathic stone formation. Urolithiasis 2014; 43 Suppl 1:77-92. [PMID: 25119505 DOI: 10.1007/s00240-014-0704-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 07/23/2014] [Indexed: 10/24/2022]
Abstract
Randall's plaque (RP) deposits seem to be consistent among the most common type of kidney stone formers, idiopathic calcium oxalate stone formers. This group forms calcium oxalate renal stones without any systemic symptoms, which contributes to the difficulty of understanding and treating this painful and recurring disease. Thus, the development of an in vitro model system to study idiopathic nephrolithiasis, beginning with RP pathogenesis, can help in identifying how plaques and subsequently stones form. One main theory of RP formation is that calcium phosphate deposits initially form in the basement membrane of the thin loops of Henle, which then fuse and spread into the interstitial tissue, and ultimately make their way across the urothelium, where upon exposure to the urine, the mineralized tissue serves as a nidus for overgrowth with calcium oxalate into a stone. Our group has found that many of the unusual morphologies found in RP and stones, such as concentrically laminated spherulites and mineralized collagenous tissue, can be reproduced in vitro using a polymer-induced liquid precursor (PILP) process, in which acidic polypeptides induce a liquid phase amorphous precursor to the mineral, yielding non-equilibrium crystal morphologies. Given that there are many acidic proteins and polysaccharides present in the renal tissue and urine, we have put forth the hypothesis that the PILP system may be involved in urolithiasis. Therefore, our goal is to develop an in vitro model system of these two stages of composite stone formation to study the role that various acidic macromolecules may play. In our initial experiments presented here, the development of "biomimetic" RP was investigated, which will then serve as a nidus for calcium oxalate overgrowth studies. To mimic the tissue environment, MatriStem(®) (ACell, Inc.), a decellularized porcine urinary bladder matrix was used, because it has both an intact epithelial basement membrane surface and a tunica propria layer, thus providing the two types of matrix constituents found associated with mineral in the early stages of RP formation. We found that when using the PILP process to mineralize this tissue matrix, the two sides led to dramatically different mineral textures, and they bore a striking resemblance to native RP, which was not seen in the tissue mineralized via the classical crystal nucleation and growth process. The interstitium side predominantly consisted of collagen-associated mineral, while the luminal side had much less mineral, which appeared to be tiny spherules embedded within the basement membrane. Although these studies are only preliminary, they support our hypothesis that kidney stones may involve non-classical crystallization pathways induced by the large variety of macromolecular species in the urinary environment. We believe that mineralization of native tissue scaffolds is useful for developing a model system of stone formation, with the ultimate goal of developing strategies to avoid RP and its detrimental consequences in stone formation, or developing therapeutic treatments to prevent or cure the disease. Supported by NIDDK grant RO1DK092311.
Collapse
|
31
|
Evan AP, Worcester EM, Coe FL, Williams J, Lingeman JE. Mechanisms of human kidney stone formation. Urolithiasis 2014; 43 Suppl 1:19-32. [PMID: 25108546 DOI: 10.1007/s00240-014-0701-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 07/23/2014] [Indexed: 01/08/2023]
Abstract
The precise mechanisms of kidney stone formation and growth are not completely known, even though human stone disease appears to be one of the oldest diseases known to medicine. With the advent of the new digital endoscope and detailed renal physiological studies performed on well phenotyped stone formers, substantial advances have been made in our knowledge of the pathogenesis of the most common type of stone former, the idiopathic calcium oxalate stone former as well as nine other stone forming groups. The observations from our group on human stone formers and those of others on model systems have suggested four entirely different pathways for kidney stone formation. Calcium oxalate stone growth over sites of Randall's plaque appear to be the primary mode of stone formation for those patients with hypercalciuria. Overgrowths off the ends of Bellini duct plugs have been noted in most stone phenotypes, do they result in a clinical stone? Micro-lith formation does occur within the lumens of dilated inner medullary collecting ducts of cystinuric stone formers and appear to be confined to this space. Lastly, cystinuric stone formers also have numerous small, oval, smooth yellow appearing calyceal stones suggestive of formation in free solution. The scientific basis for each of these four modes of stone formation are reviewed and used to explore novel research opportunities.
Collapse
Affiliation(s)
- Andrew P Evan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 5055, Indianapolis, IN, 46220, USA,
| | | | | | | | | |
Collapse
|
32
|
Genetic modulation of nephrocalcinosis in mouse models of ectopic mineralization: the Abcc6(tm1Jfk) and Enpp1(asj) mutant mice. J Transl Med 2014; 94:623-32. [PMID: 24732453 PMCID: PMC4039617 DOI: 10.1038/labinvest.2014.52] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/12/2014] [Accepted: 02/27/2014] [Indexed: 11/09/2022] Open
Abstract
Ectopic mineralization of renal tissues in nephrocalcinosis is a complex, multifactorial process. The purpose of this study was to examine the role of genetic modulation and the role of diet in nephrocalcinosis using two established mouse models of ectopic mineralization, Abcc6(tm1Jfk) and Enpp1(asj) mice, which serve as models for pseudoxanthoma elasticum and generalized arterial calcification of infancy, two heritable disorders, respectively. These mutant mice, when on standard rodent diet, develop nephrocalcinosis only at a very late age. In contrast, when placed on an 'acceleration diet' composed of increased phosphate and reduced magnesium content, they showed extensive mineralization of the kidneys affecting primarily the medullary tubules as well as arcuate and renal arteries, as examined by histopathology and quantitated by chemical assay for calcium. Mineralization could also be detected noninvasively by micro computed tomography. Whereas the heterozygous mice did not develop nephrocalcinosis, compound heterozygous mice carrying both mutant alleles, Abcc6(tm1Jfk/+) and Enpp1(+/asj), developed ectopic mineralization similar to that noted in homozygous mice for either gene, indicating that deletion of one Abcc6 allele along with Enpp1 haploinsufficiency resulted in renal mineralization. Thus, synergistic genetic defects in the complex mineralization/antimineralization network can profoundly modulate the degree of ectopic mineralization in nephrocalcinosis.
Collapse
|
33
|
Evan AP, Lingeman JE, Worcester EM, Sommer AJ, Phillips CL, Williams JC, Coe FL. Contrasting histopathology and crystal deposits in kidneys of idiopathic stone formers who produce hydroxy apatite, brushite, or calcium oxalate stones. Anat Rec (Hoboken) 2014; 297:731-48. [PMID: 24478243 DOI: 10.1002/ar.22881] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 12/16/2013] [Indexed: 12/11/2022]
Abstract
Our previous work has shown that stone formers who form calcium phosphate (CaP) stones that contain any brushite (BRSF) have a distinctive renal histopathology and surgical anatomy when compared with idiopathic calcium oxalate stone formers (ICSF). Here we report on another group of idiopathic CaP stone formers, those forming stone containing primarily hydroxyapatite, in order to clarify in what ways their pathology differs from BRSF and ICSF. Eleven hydroxyapatite stone formers (HASF) (2 males, 9 females) were studied using intra-operative digital photography and biopsy of papillary and cortical regions to measure tissue changes associated with stone formation. Our main finding is that HASF and BRSF differ significantly from each other and that both differ greatly from ICSF. Both BRSF and ICSF patients have significant levels of Randall's plaque compared with HASF. Intra-tubular deposit number is greater in HASF than BRSF and nonexistent in ICSF while deposit size is smaller in HASF than BRSF. Cortical pathology is distinctly greater in BRSF than HASF. Four attached stones were observed in HASF, three in 25 BRSF and 5-10 per ICSF patient. HASF and BRSF differ clinically in that both have higher average urine pH, supersaturation of CaP, and calcium excretion than ICSF. Our work suggests that HASF and BRSF are two distinct and separate diseases and both differ greatly from ICSF.
Collapse
Affiliation(s)
- Andrew P Evan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Urology, International Kidney Stone Institute, Methodist Hospital, Indianapolis, Indiana
| | | | | | | | | | | | | |
Collapse
|
34
|
Giardina S, Scilironi C, Michelotti A, Samuele A, Borella F, Daglia M, Marzatico F. In vitro anti-inflammatory activity of selected oxalate-degrading probiotic bacteria: potential applications in the prevention and treatment of hyperoxaluria. J Food Sci 2014; 79:M384-90. [PMID: 24471378 DOI: 10.1111/1750-3841.12344] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 11/25/2013] [Indexed: 01/06/2023]
Abstract
UNLABELLED Oxalate (Ox) is a very common component of the human diet, capable to collect in the renal tissue and bind calcium to form calcium oxalate (CaOx) crystals. A supersaturation of CaOx crystal may cause nephrocalcinosis and nephrolithiasis. The inflammation derived from the CaOx crystal accumulation, together with innate or secondary renal alterations, could strongly affect the renal function. In this case a consumption of probiotics with either oxalate-degrading activity at intestinal level and systemic anti-inflammatory activity could be an alternative approach to treat the subjects with excess of urinary oxalate excretion. 11 strains of lactic acid bacteria (Lactobacilli and Bifidobacteria), already included in the list of bacteria safe for the human use, were investigated for their capability to degrade oxalate by mean of RP-HPLC-UV method and modulate inflammation in an in vitro model system based on peripheral blood mononuclear cells. Four promising bacterial strains (Lactobacillus plantarum PBS067, Lactobacillus acidophilus LA-14, Bifidobacterium breve PBS077, Bifidobacterium longum PBS078) were identified as innovative biological tools for the prevention and the therapeutic treatment of hyperoxaluria and the inflammatory events associated to the Ox accumulation. PRACTICAL APPLICATION The oxalate-degrading activity of some probiotics and their capability to modulate the release of inflammation mediators could be exploited as a new nutraceutical and therapeutic approach for the treatment of oxalate accumulation and the related inflammatory state.
Collapse
Affiliation(s)
- Silvana Giardina
- Farcoderm Srl, Via Angelini, 21-27028, San Martino Siccomario (PV), Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Worcester EM, Evan AP, Coe FL, Lingeman JE, Krambeck A, Sommers A, Phillips CL, Milliner D. A test of the hypothesis that oxalate secretion produces proximal tubule crystallization in primary hyperoxaluria type I. Am J Physiol Renal Physiol 2013; 305:F1574-84. [PMID: 24089413 DOI: 10.1152/ajprenal.00382.2013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The sequence of events by which primary hyperoxaluria type 1 (PH1) causes renal failure is unclear. We hypothesize that proximal tubule (PT) is vulnerable because oxalate secretion raises calcium oxalate (CaOx) supersaturation (SS) there, leading to crystal formation and cellular injury. We studied cortical and papillary biopsies from two PH1 patients with preserved renal function, and seven native kidneys removed from four patients at the time of transplant, after short-term (2) or longer term (2) dialysis. In these patients, and another five PH1 patients without renal failure, we calculated oxalate secretion, and estimated PT CaOx SS. Plasma oxalate was elevated in all PH1 patients and inverse to creatinine clearance. Renal secretion of oxalate was present in all PH1 but rare in controls. PT CaOx SS was >1 in all nonpyridoxine-responsive PH1 before transplant and most marked in patients who developed end stage renal disease (ESRD). PT from PH1 with preserved renal function had birefringent crystals, confirming the presence of CaOx SS, but had no evidence of cortical inflammation or scarring by histopathology or hyaluronan staining. PH1 with short ESRD showed CaOx deposition and hyaluronan staining particularly at the corticomedullary junction in distal PT while cortical collecting ducts were spared. Longer ESRD showed widespread cortical CaOx, and in both groups papillary tissue had marked intratubular CaOx deposits and fibrosis. CaOx SS in PT causes CaOx crystal formation, and CaOx deposition in distal PT appears to be associated with ESRD. Minimizing PT CaOx SS may be important for preserving renal function in PH1.
Collapse
Affiliation(s)
- Elaine M Worcester
- Nephrology Section, MC5100, Univ. of Chicago, School of Medicine, 5841 South Maryland Ave., Chicago, IL 60637.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
NADPH oxidase as a therapeutic target for oxalate induced injury in kidneys. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:462361. [PMID: 23840917 PMCID: PMC3690252 DOI: 10.1155/2013/462361] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023]
Abstract
A major role of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes is to catalyze the production of superoxides and other reactive oxygen species (ROS). These ROS, in turn, play a key role as messengers in cell signal transduction and cell cycling, but when they are produced in excess they can lead to oxidative stress (OS). Oxidative stress in the kidneys is now considered a major cause of renal injury and inflammation, giving rise to a variety of pathological disorders. In this review, we discuss the putative role of oxalate in producing oxidative stress via the production of reactive oxygen species by isoforms of NADPH oxidases expressed in different cellular locations of the kidneys. Most renal cells produce ROS, and recent data indicate a direct correlation between upregulated gene expressions of NADPH oxidase, ROS, and inflammation. Renal tissue expression of multiple NADPH oxidase isoforms most likely will impact the future use of different antioxidants and NADPH oxidase inhibitors to minimize OS and renal tissue injury in hyperoxaluria-induced kidney stone disease.
Collapse
|
37
|
Amer H, Griffin MD, Stegall MD, Cosio FG, Park WD, Kremers WK, Heilman RL, Mazur MJ, Hamawi K, Larson TS, Kumar R. Oral paricalcitol reduces the prevalence of posttransplant hyperparathyroidism: results of an open label randomized trial. Am J Transplant 2013; 13:1576-85. [PMID: 23601186 DOI: 10.1111/ajt.12227] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 02/12/2013] [Accepted: 02/15/2013] [Indexed: 01/25/2023]
Abstract
Postkidney transplant hyperparathyroidism is a significant problem. Vitamin D receptor agonists are known to suppress parathyroid hormone (PTH) secretion. We examined the effect of oral paricalcitol on posttransplant secondary hyperparathyroidism by conducting an open label randomized trial in which 100 incident kidney transplant recipients were randomized 1:1 to receive oral paricalcitol, 2 μg per day, for the first year posttransplant or no additional therapy. Serial measurements of serum PTH, calcium and bone alkaline phosphatase, 24-h urine calcium and bone density were performed. The primary endpoint was the frequency of hyperparathyroidism 1-year posttransplant. Eighty-seven patients completed the trial. One-year posttransplant, 29% of paricalcitol-treated subjects had hyperparathyroidism compared with 63% of untreated patients (p = 0.0005). Calcium supplementation was discontinued in two control and 15 treatment patients due to mild hypercalcemia or hypercalcuria. Paricalcitol was discontinued in four patients due to hypercalcuria/hypercalcemia and in one for preference. Two subjects required decreasing the dose of paricalcitol to 1 μg daily. Hypercalcemia was asymptomatic and reversible. Incidence of acute rejection, BK nephropathy and renal function at 1 year were similar between groups. Moderate renal allograft fibrosis was reduced in treated patients. Oral paricalcitol is effective in decreasing posttransplant hyperparathyroidism and may have beneficial effects on renal allograft histology.
Collapse
Affiliation(s)
- H Amer
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Okabe C, Borges RL, de Almeida DC, Fanelli C, Barlette GP, Machado FG, Arias SCA, Malheiros DMAC, Camara NOS, Zatz R, Fujihara CK. NF-κB activation mediates crystal translocation and interstitial inflammation in adenine overload nephropathy. Am J Physiol Renal Physiol 2013; 305:F155-63. [PMID: 23657853 DOI: 10.1152/ajprenal.00491.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenine overload promotes intratubular crystal precipitation and interstitial nephritis. We showed recently that these abnormalities are strongly attenuated in mice knockout for Toll-like receptors-2, -4, MyD88, ASC, or caspase-1. We now investigated whether NF-κB activation also plays a pathogenic role in this model. Adult male Munich-Wistar rats were distributed among three groups: C (n = 17), receiving standard chow; ADE (n = 17), given adenine in the chow at 0.7% for 1 wk and 0.5% for 2 wk; and ADE + pyrrolidine dithiocarbamate (PDTC; n = 14), receiving adenine as above and the NF-κB inhibitor PDTC (120 mg·kg⁻¹·day⁻¹ in the drinking water). After 3 wk, widespread crystal deposition was seen in tubular lumina and in the renal interstitium, along with granuloma formation, collagen accumulation, intense tubulointerstitial proliferation, and increased interstitial expression of inflammatory mediators. Part of the crystals were segregated from tubular lumina by a newly formed cell layer and, at more advanced stages, appeared to be extruded to the interstitium. p65 nuclear translocation and IKK-α increased abundance indicated activation of the NF-κB system. PDTC treatment prevented p65 migration and normalized IKK-α, limited crystal shift to the interstitium, and strongly attenuated interstitial fibrosis/inflammation. These findings indicate that the complex inflammatory phenomena associated with this model depend, at least in part, on NF-κB activation, and suggest that the NF-κB system may become a therapeutic target in the treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Cristiene Okabe
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chiang WF, Yan MT, Wu TJ, Lin SH. A hypokalaemic woman with nephrocalcinosis: rebirth of old knowledge. Ann Clin Biochem 2013; 50:176-9. [PMID: 23440542 DOI: 10.1177/0004563212474551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The coexistence of hypokalaemia and nephrocalcinosis poses a challenge in rapid diagnosis and appropriate management. We describe a 38-year-old woman who presented with thirst, intermittent carpopedal spasm, paresthaesia of both hands and progressive weakness of lower extremities for two years. She had a history of chronic hypokalaemia of unknown cause with intermittent potassium supplementation for 7-8 y and bilateral nephrocalcinosis notable for one year. She denied vomiting, diarrhoea or use of laxatives, alcohol or diuretics. Her blood pressure was normal. Laboratory investigations showed hypokalaemia (2.7 mmol/L) and metabolic alkalosis (HCO3(-) 32.6 mmol/L, pH 7.46). Two random urine samples both showed a consistently high urine K(+) excretion but with excretion rates of Na(+), Cl(-) and divalent cations which were high in one sample but not the other. Ingestion of furosemide 120 mg daily for body image for 7-8 y was uncovered. With furosemide cessation and potassium supplementation, her hypokalaemia with neuromuscular symptoms was corrected but nephrocalcinosis persisted. Surreptitious use of diuretics for various purposes should be kept in mind as an important cause of hypokalaemia and/or nephrocalcinosis. Measurement of electrolyte concentrations in at least two random urine samples is warranted to distinguish it from true renal tubular disorders and extrarenal causes.
Collapse
Affiliation(s)
- Wen-Fang Chiang
- Department of Medicine, Armed Forces Taoyuan General Hospital, Taoyuan 325, Taiwan
| | | | | | | |
Collapse
|
40
|
Nesterova G, Malicdan MC, Yasuda K, Sakaki T, Vilboux T, Ciccone C, Horst R, Huang Y, Golas G, Introne W, Huizing M, Adams D, Boerkoel CF, Collins MT, Gahl WA. 1,25-(OH)2D-24 Hydroxylase (CYP24A1) Deficiency as a Cause of Nephrolithiasis. Clin J Am Soc Nephrol 2013; 8:649-57. [PMID: 23293122 DOI: 10.2215/cjn.05360512] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES Elevated serum vitamin D with hypercalciuria can result in nephrocalcinosis and nephrolithiasis. This study evaluated the cause of excess 1,25-dihydroxycholecalciferol (1α,25(OH)2D3) in the development of those disorders in two individuals. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Two patients with elevated vitamin D levels and nephrocalcinosis or nephrolithiasis were investigated at the National Institutes of Health (NIH) Clinical Center and the NIH Undiagnosed Diseases Program, by measuring calcium, phosphate, and vitamin D metabolites, and by performing CYP24A1 mutation analysis. RESULTS Both patients exhibited hypercalciuria, hypercalcemia, low parathyroid hormone, elevated vitamin D (1α,25(OH)2D3), normal 25-OHD3, decreased 24,25(OH)2D, and undetectable activity of 1,25(OH)2D-24-hydroxylase (CYP24A1), the enzyme that inactivates 1α,25(OH)2D3. Both patients had bi-allelic mutations in CYP24A1 leading to loss of function of this enzyme. On the basis of dbSNP data, the frequency of predicted deleterious bi-allelic CYP24A1 variants in the general population is estimated to be as high as 4%-20%. CONCLUSIONS The results of this study show that 1,25(OH)2D-24-hydroxylase deficiency due to bi-allelic mutations in CYP24A1 causes elevated serum vitamin D, hypercalciuria, nephrocalcinosis, and renal stones.
Collapse
Affiliation(s)
- Galina Nesterova
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Miyazawa K, Takahashi Y, Morita N, Moriyama MT, Kosaka T, Nishio M, Yoshimoto T, Suzuki K. Cyclooxygenase 2 and prostaglandin E2 regulate the attachment of calcium oxalate crystals to renal epithelial cells. Int J Urol 2012; 19:936-43. [PMID: 22640700 DOI: 10.1111/j.1442-2042.2012.03060.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To determine the roles of endogenous cyclooxygenase 2 and prostaglandin E(2) in crystal-cell binding, which is considered to be an important step in the development of intratubular nephrocalcinosis. METHODS An expression plasmid for human cyclooxygenase 2 was introduced into Madin-Darby canine kidney cells using the lipofection method. Cyclooxygenase activity was measured using thin-layer chromatography, and the prostaglandin E(2) concentration was determined with an enzyme immunoassay. In addition, crystal attachment was evaluated with a liquid scintillation counter using [(14)C] calcium oxalate monohydrate crystals, and immunohistochemistry and an enzyme immunoassay were used to analyze and quantify the expression of hyaluronan, a crystal-binding molecule. RESULTS Cyclooxygenase 2-overexpressing Madin-Darby canine kidney cells produced about 10-fold more prostaglandin E(2) than wild-type Madin-Darby canine kidney cells, and their hyaluronan production was also upregulated. The attachment of calcium oxalate monohydrate crystals to cyclooxygenase 2-overexpressing Madin-Darby canine kidney cells was significantly reduced compared with their attachment to wild-type and mock-transfected Madin-Darby canine kidney cells. Pre-incubation of the cyclooxygenase 2-overexpressing cells, as well as the mock-transfected and wild-type cells with the cyclooxygenase 2 selective inhibitor etodolac, increased the cellular attachment of calcium oxalate monohydrate crystals in a dose-dependent manner. CONCLUSIONS These findings suggest that cyclooxygenase 2 expression and the resultant increase in endogenous prostaglandin E(2), leading to increased hyaluronan production, help to prevent nephrocalcinosis by inhibiting the attachment of calcium oxalate monohydrate crystals to the surface of renal epithelial cells.
Collapse
Affiliation(s)
- Katsuhito Miyazawa
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Damasceno Zanuto AC, Bueno T, Alvavares Delfino VD, Jacob Mocelin A. Nephrocalcinosis in a patient with Sjögren's syndrome/systemic lupus erythematosus. Rev Assoc Med Bras (1992) 2012. [DOI: 10.1016/s0104-4230(12)70194-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
43
|
Damasceno Zanuto AC, Bueno T, Alvares Delfino VD, Mocelin AJ. Nephrocalcinosis in a patient with Sjögren's syndrome/systemic lupus erythematosus. Rev Assoc Med Bras (1992) 2012. [DOI: 10.1590/s0104-42302012000300004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
44
|
Koçak G, Koçak E, Azak A, Unverdi H. An unusual cause of acute renal failure in a patient with surgical hypoparathyroidism: nephrocalcinosis. Endocrine 2012; 41:162-3. [PMID: 22083502 DOI: 10.1007/s12020-011-9561-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Evenepoel P, Sprangers B, Lerut E, Bammens B, Claes K, Kuypers D, Meijers B, Vanrenterghem Y. Mineral metabolism in renal transplant recipients discontinuing cinacalcet at the time of transplantation: a prospective observational study. Clin Transplant 2011; 26:393-402. [PMID: 22007637 DOI: 10.1111/j.1399-0012.2011.01524.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The calcimimetic cinacalcet is approved for treating secondary hyperparathyroidism in patients with chronic kidney disease on dialysis. Biochemical profiles and clinical outcomes in patients discontinuing cinacalcet at the time of transplantation are scarce. METHODS We performed a prospective observational cohort study, including 303 incident renal transplant recipients, of whom 21 were on cinacalcet treatment at the time of transplantation. Parameters of mineral metabolism and incidence of parathyroidectomy and nephrocalcinosis in patients discontinuing cinacalcet at the time of transplantation patients ("cinacalcet +") were compared to cinacalcet-naïve patients ("cinacalcet -"). Mean follow-up was 35.6 ± 15.8 months. RESULTS At the time of transplantation, parameters of mineral metabolism were similar in both groups. Conversely, at month 3, serum ionized calcium (p = 0.0007), calcitriol (p = 0.02), biointact parathyroid hormone (p = 0.06) levels and urinary fractional excretion of phosphorus (p = 0.06) were higher, while serum phosphorus levels (p = 0.06) were lower in "cinacalcet +." Analysis based on matching at the time of initiation showed that the course of post-transplant mineral metabolism in cinacalcet-treated patients (median treatment period 12.5 months) vs. cinacalcet-naïve patients was identical. "Cinacalcet +" patients are characterized by a high-incidence proportion of both post-transplant nephrocalcinosis (45% at month 3) and parathyroidectomy (28.6%). No difference in renal function was observed between "cinacalcet +" and "cinacalcet-" patients. CONCLUSION Cinacalcet does not affect the course of secondary hyperparathyroidism in patients awaiting kidney transplantation. Biochemical profiles and a high parathyroidectomy rate suggest rebound hyperparathyroidism in renal transplant recipients discontinuing cinacalcet at the time of transplantation, which may be related to the short exposure time specific to this population. Risk/benefit studies are urgently required to define the role of continued calcimimetic treatment in renal transplant recipients and to determine the optimal treatment of secondary hyperparathyroidism in patients listed for transplantation.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Division of Nephrology, Department of Medicine, University Hospitals Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Wong PN, Chan ANH, Lo KY, Wong YY, Mak SK, Wong AKM. Kidney allograft failure due to acute phosphate nephropathy associated with severe secondary hyperparathyroidism. NDT Plus 2011; 4:324-6. [PMID: 25984179 PMCID: PMC4421720 DOI: 10.1093/ndtplus/sfr078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 05/31/2011] [Indexed: 11/14/2022] Open
Abstract
Intratubular calcification is a common finding in renal allografts. However, possible harmful effect of this calcification is not well recognized, and allograft failure purely due to this condition has not been reported. We report a kidney transplant recipient who suffered from severe secondary hyperparathyroidism and unexplained early allograft failure. A diagnosis of acute phosphate nephropathy was made subsequently based on serial allograft biopsy findings. This case calls for a high index of suspicion to look for this rare cause of allograft dysfunction among high-risk patients. It also highlights the importance of good calcium–phosphate control before renal transplantation.
Collapse
Affiliation(s)
- Ping-Nam Wong
- Renal Unit, Department of Medicine & Geriatrics, Kwong Wah Hospital, Hong Kong SAR, China
| | - Alice N H Chan
- Department of Pathology, Kwong Wah Hospital, Hong Kong SAR, China
| | - Kin-Yee Lo
- Renal Unit, Department of Medicine & Geriatrics, Kwong Wah Hospital, Hong Kong SAR, China
| | - Yuk-Yi Wong
- Renal Unit, Department of Medicine & Geriatrics, Kwong Wah Hospital, Hong Kong SAR, China
| | - Siu-Ka Mak
- Renal Unit, Department of Medicine & Geriatrics, Kwong Wah Hospital, Hong Kong SAR, China
| | - Andrew K M Wong
- Renal Unit, Department of Medicine & Geriatrics, Kwong Wah Hospital, Hong Kong SAR, China
| |
Collapse
|
47
|
Sampaio MS, Molnar MZ, Kovesdy CP, Mehrotra R, Mucsi I, Sim JJ, Krishnan M, Nissenson AR, Kalantar-Zadeh K. Association of pretransplant serum phosphorus with posttransplant outcomes. Clin J Am Soc Nephrol 2011; 6:2712-21. [PMID: 21959597 DOI: 10.2215/cjn.06190611] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Serum phosphorus levels are associated with mortality, cardiovascular disease, and renal function loss in individuals with and without chronic kidney disease. The association of pretransplant serum phosphorus levels with transplant outcomes is not clear. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Data of the Scientific Registry of Transplant Recipients (SRTR) up to June 2007 were linked to the database (2001 through 2006) of one of the U.S.-based large dialysis organizations (DaVita). The selected 9384 primary kidney recipients were divided into five groups according to pretransplant serum phosphorus levels (mg/dl): <3.5, 3.5 to <5.5 (reference group), 5.5 to <7.5, 7.5 to <9.5, and ≥9.5. Unadjusted and multivariate adjusted risks for transplant outcomes were compared. RESULTS Patients were 48 ± 14 years old and included 37% women and 27% African Americans. After multivariate adjustment, all-cause and cardiovascular death hazard ratios were 2.44 (95% confidence interval: 1.28 to 4.65) and 3.63 (1.13 to 11.64), respectively, in recipients in the ≥9.5 group; allograft loss hazard ratios were 1.42 (1.04 to 1.95) and 2.36 (1.33 to 4.17) in recipients with 7.5 to >9.5 and ≥9.5, respectively. No significant association with delayed graft function was found. CONCLUSIONS Pretransplant phosphorus levels 7.5 to <9.5 mg/dl and ≥9.5 mg/dl were associated with increased risk of functional graft failure and increased risk of all-cause and cardiovascular deaths, respectively, when compared with 3.5 to <5.5 mg/dl. Additional studies are needed to examine whether more aggressive control of pretransplant serum phosphorus may improve posttransplant outcomes.
Collapse
Affiliation(s)
- Marcelo S Sampaio
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90509-2910, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The incidence of adult urolithiasis has increased significantly in industrialized countries over the past decades. Sound incidence rates are not available for children, nor are they known for nephrocalcinosis, which can appear as a single entity or together with urolithiasis. In contrast to the adult kidney stone patient, where environmental factors are the main cause, genetic and/or metabolic disorders are the main reason for childhood nephrocalcinosis and urolithiasis. While hypercalciuria is considered to be the most frequent risk factor, several other metabolic disorders such as hypocitraturia or hyperoxaluria, as well as a variety of renal tubular diseases, e.g., Dent's disease or renal tubular acidosis, have to be ruled out by urine and/or blood analysis. Associated symptoms such as growth retardation, intestinal absorption, or bone demineralization should be evaluated for diagnostic and therapeutic purposes. Preterm infants are a special risk population with a high incidence of nephrocalcinosis arising from immature kidney, medication, and hypocitraturia. In children, concise evaluation will reveal an underlying pathomechanism in >75% of patients. Early treatment reducing urinary saturation of the soluble by increasing fluid intake and by providing crystallization inhibitors, as well as disease-specific medication, are mandatory to prevent recurrent kidney stones and/or progressive nephrocalcinosis, and consequently deterioration of renal function.
Collapse
|
49
|
Thurgood LA, Sørensen ES, Ryall RL. The effect of intracrystalline and surface-bound osteopontin on the degradation and dissolution of calcium oxalate dihydrate crystals in MDCKII cells. ACTA ACUST UNITED AC 2011; 40:1-15. [PMID: 21932131 DOI: 10.1007/s00240-011-0423-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 08/22/2011] [Indexed: 01/12/2023]
Abstract
In vivo, urinary crystals are associated with proteins located within the mineral bulk as well as upon their surfaces. Proteins incarcerated within the mineral phase of retained crystals could act as a defence against urolithiasis by rendering them more vulnerable to destruction by intracellular and interstitial proteases. The aim of this study was to examine the effects of intracrystalline and surface-bound osteopontin (OPN) on the degradation and dissolution of urinary calcium oxalate dihydrate (COD) crystals in cultured Madin Darby canine kidney (MDCK) cells. [(14)C]-oxalate-labelled COD crystals with intracrystalline (IC), surface-bound (SB) and IC + SB OPN, were generated from ultrafiltered (UF) urine containing 0, 1 and 5 mg/L human milk OPN and incubated with MDCKII cells, using UF urine as the binding medium. Crystal size and degradation were assessed using field emission scanning electron microscopy (FESEM) and dissolution was quantified by the release of radioactivity into the culture medium. Crystal size decreased directly with OPN concentration. FESEM examination indicated that crystals covered with SB OPN were more resistant to cellular degradation than those containing IC OPN, whose degree of disruption appeared to be related to OPN concentration. Whether bound to the crystal surface or incarcerated within the mineral interior, OPN inhibited crystal dissolution in direct proportion to its concentration. Under physiological conditions OPN may routinely protect against stone formation by inhibiting the growth of COD crystals, which would encourage their excretion in urine and thereby perhaps partly explain why, compared with calcium oxalate monohydrate crystals, COD crystals are more prevalent in urine, but less common in kidney stones.
Collapse
Affiliation(s)
- Lauren A Thurgood
- Urology Unit, Department of Surgery, Flinders Medical Centre, Flinders University, Bedford Park, SA 5042, Australia
| | | | | |
Collapse
|
50
|
Thurgood LA, Sørensen ES, Ryall RL. The effect of intracrystalline and surface-bound osteopontin on the attachment of calcium oxalate dihydrate crystals to Madin-Darby canine kidney (MDCK) cells in ultrafiltered human urine. BJU Int 2011; 109:1100-9. [DOI: 10.1111/j.1464-410x.2011.10530.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|