1
|
Yang W, Zhao T, Chen X, Wang S, Wang Y, Su T. Determinants and impact of calcium oxalate crystal deposition on renal outcomes in acute kidney injury patients. Ren Fail 2024; 46:2334396. [PMID: 38570195 PMCID: PMC10993744 DOI: 10.1080/0886022x.2024.2334396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVES Calcium oxalate (CaOx) crystal deposition in acute kidney injury (AKI) patients is under recognized but impacts renal outcomes. This study investigates its determinants and effects. METHODS We studied 814 AKI patients with native kidney biopsies from 2011 to 2020, identifying CaOx crystal deposition severity (mild: <5, moderate: 5-10, severe: >10 crystals per section). We assessed factors like urinary oxalate, citrate, urate, electrolytes, pH, tubular calcification index, and SLC26A6 expression, comparing them with creatinine-matched AKI controls without oxalosis. We analyzed how these factors relate to CaOx severity and their impact on renal recovery (eGFR < 15 mL/min/1.73 m2 at 3-month follow-up). RESULTS CaOx crystal deposition was found in 3.9% of the AKI cohort (32 cases), with 72% due to nephrotoxic medication-induced tubulointerstitial nephritis. Diuretic use, higher urinary oxalate-to-citrate ratio induced by hypocitraturia, and tubular calcification index were significant contributors to moderate and/or severe CaOx deposition. Poor baseline renal function, low urinary chloride, high uric acid and urea nitrogen, tubular SLC26A6 overexpression, and glomerular sclerosis were also associated with moderate-to-severe CaOx deposition. Kidney recovery was delayed, with 43.8%, 31.2%, and 18.8% of patients having eGFR < 15 mL/min/1.73 m2 at 4, 12, and 24-week post-injury. Poor outcomes were linked to high urinary α1-microglobulin-to-creatinine (α1-MG/C) ratios and active tubular injury scores. Univariate analysis showed a strong link between this ratio and poor renal outcomes, independent of oxalosis severity. CONCLUSIONS In AKI, CaOx deposition is common despite declining GFR. Factors worsening tubular injury, not just oxalate-to-citrate ratios, are key to understanding impaired renal recovery.
Collapse
Affiliation(s)
- Weiwei Yang
- Department of medicine, Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, PR China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, PR China
| | - Tao Zhao
- Department of medicine, Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, PR China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, PR China
| | - Xuejing Chen
- Department of medicine, Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, PR China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, PR China
| | - Suxia Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, PR China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Yu Wang
- Department of medicine, Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, PR China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, PR China
| | - Tao Su
- Department of medicine, Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, PR China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, PR China
| |
Collapse
|
2
|
Aggarwal N, Singh G, Panda HS, Panda JJ. Unravelling the potential of L-carnosine analog-based nano-assemblies as pH-responsive therapeutics in treating glioma: an in vitro perspective. J Mater Chem B 2024; 12:10665-10681. [PMID: 39314035 DOI: 10.1039/d4tb01262c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Self-assembled small peptide-based nanoparticles (NPs) constitute a major section of the biomimetic smart NPs owing to their excellent compatibility and minimal adverse effects in the biological system. Here, we have designed a modified L-carnosine dipeptide analog, "Fmoc-β-Ala-L-His-(Trt)-o-methyl formate", which was assembled along with a modified single amino acid, Fmoc-Arg-(Pbf)-OH and zinc ions to form stable and mono-dispersed L-carnosine analog NPs (CaNPs) with inherent anti-cancer properties. Furthermore, the CaNPs demonstrated an average size of ∼200 nm, making them suitable to invade the tumor site by following the enhanced permeability and retention (EPR) effect. Our studies depicted a remarkable cancer cell killing ability of the NPs of ∼82% in C6 glioma cells. Thereafter, cellular investigations were performed in C6 cells to analyze the influence of the NPs on cellular cytoskeleton integrity by using a phalloidin assay and anti-cancer efficacy by using calcein AM/PI, and an apoptosis assay further indicated their anti-cancer effect. Additionally, the NPs negatively impacted the ability of C6 cells to migrate across a premade scratch (∼44% wound closure) demonstrating their tendency to halt cancer cell migration and metastasis. Also, our NPs depicted ∼19.51 ± 0.17% permeability across the bEnd.3 transwell model establishing their BBB penetrability. Collectively, our results could positively implicate the successful anti-cancer potential of the minimalistic, biologically compliant, L-carnosine analog (Ca)-based nanostructures in glioma.
Collapse
Affiliation(s)
- Nidhi Aggarwal
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Mohali, Punjab 140306, India.
| | - Gurjot Singh
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Mohali, Punjab 140306, India.
| | - Himanshu Sekhar Panda
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Mohali, Punjab 140306, India.
| | - Jiban Jyoti Panda
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Mohali, Punjab 140306, India.
| |
Collapse
|
3
|
Heng BL, Wu FY, Tong XY, Zou GJ, Ouyang JM. Corn Silk Polysaccharide Reduces the Risk of Kidney Stone Formation by Reducing Oxidative Stress and Inhibiting COM Crystal Adhesion and Aggregation. ACS OMEGA 2024; 9:19236-19249. [PMID: 38708219 PMCID: PMC11064203 DOI: 10.1021/acsomega.4c00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 05/07/2024]
Abstract
The aim of this study is to explore the inhibition of nanocalcium oxalate monohydrate (nano-COM) crystal adhesion and aggregation on the HK-2 cell surface after the protection of corn silk polysaccharides (CSPs) and the effect of carboxyl group (-COOH) content and polysaccharide concentration. METHOD HK-2 cells were damaged by 100 nm COM crystals to build an injury model. The cells were protected by CSPs with -COOH contents of 3.92% (CSP0) and 16.38% (CCSP3), respectively. The changes in the biochemical indexes of HK-2 cells and the difference in adhesion amount and aggregation degree of nano-COM on the cell surface before and after CSP protection were detected. RESULTS CSP0 and CCSP3 protection can obviously inhibit HK-2 cell damage caused by nano-COM crystals, restore cytoskeleton morphology, reduce intracellular ROS level, inhibit phosphoserine eversion, restore the polarity of the mitochondrial membrane potential, normalize the cell cycle process, and reduce the expression of adhesion molecules, OPN, Annexin A1, HSP90, HAS3, and CD44 on the cell surface. Finally, the adhesion and aggregation of nano-COM crystals on the cell surface were effectively inhibited. The carboxymethylated CSP3 exhibited a higher protective effect on cells than the original CSP0, and cell viability was further improved with the increase in polysaccharide concentration. CONCLUSIONS CSPs can protect HK-2 cells from calcium oxalate crystal damage and effectively reduce the adhesion and aggregation of nano-COM crystals on the cell surface, which is conducive to inhibiting the formation of calcium oxalate kidney stones.
Collapse
Affiliation(s)
- Bao-Li Heng
- Yingde
Center, Institute of Kidney Surgery, Jinan
University, Guangdong 510000, China
- Department
of Urology, People’s Hospital of
Yingde City, Yingde 513000, China
| | - Fan-Yu Wu
- Yingde
Center, Institute of Kidney Surgery, Jinan
University, Guangdong 510000, China
- Department
of Urology, People’s Hospital of
Yingde City, Yingde 513000, China
| | - Xin-Yi Tong
- Institute
of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Guo-Jun Zou
- Institute
of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Institute
of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| |
Collapse
|
4
|
Chmiel JA, Stuivenberg GA, Al KF, Akouris PP, Razvi H, Burton JP, Bjazevic J. Vitamins as regulators of calcium-containing kidney stones - new perspectives on the role of the gut microbiome. Nat Rev Urol 2023; 20:615-637. [PMID: 37161031 PMCID: PMC10169205 DOI: 10.1038/s41585-023-00768-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 05/11/2023]
Abstract
Calcium-based kidney stone disease is a highly prevalent and morbid condition, with an often complicated and multifactorial aetiology. An abundance of research on the role of specific vitamins (B6, C and D) in stone formation exists, but no consensus has been reached on how these vitamins influence stone disease. As a consequence of emerging research on the role of the gut microbiota in urolithiasis, previous notions on the contribution of these vitamins to urolithiasis are being reconsidered in the field, and investigation into previously overlooked vitamins (A, E and K) was expanded. Understanding how the microbiota influences host vitamin regulation could help to determine the role of vitamins in stone disease.
Collapse
Affiliation(s)
- John A Chmiel
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Gerrit A Stuivenberg
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Kait F Al
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Polycronis P Akouris
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Hassan Razvi
- Division of Urology, Department of Surgery, Western University, London, Ontario, Canada
| | - Jeremy P Burton
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
- Division of Urology, Department of Surgery, Western University, London, Ontario, Canada
| | - Jennifer Bjazevic
- Division of Urology, Department of Surgery, Western University, London, Ontario, Canada.
| |
Collapse
|
5
|
Antioxidant Activities and Cytotoxicity of the Regulated Calcium Oxalate Crystals on HK-2 Cells of Polysaccharides from Gracilaria lemaneiformis with Different Molecular Weights. Foods 2023; 12:foods12051031. [PMID: 36900548 PMCID: PMC10001015 DOI: 10.3390/foods12051031] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 03/06/2023] Open
Abstract
The antioxidant activities of seven degraded products (GLPs) with different molecular weights (Mw) of polysaccharides from Gracilaria lemaneiformis were compared. The Mw of GLP1-GLP7 were 106, 49.6, 10.5, 6.14, 5.06, 3.71 and 2.42 kDa, respectively. The results show that GLP2 with Mw = 49.6 kDa had the strongest scavenging capacity for hydroxyl radical, DPPH radical, ABTS radical and reducing power. When Mw < 49.6 kDa, the antioxidant activity of GLPs increased with the increase in Mw, but when Mw increased to 106 kDa, their antioxidant activity decreased. However, the ability of GLPs to chelate Fe2+ ions increased with the decrease in polysaccharide Mw, which was attributed to the fact that the polysaccharide active groups (-OSO3- and -COOH) were easier to expose, and the steric hindrance was smaller when GLPs chelated with Fe2+. The effects of GLP1, GLP3, GLP5 and GLP7 on the crystal growth of calcium oxalate (CaOx) were studied using XRD, FT-IR, Zeta potential and thermogravimetric analysis. Four kinds of GLPs could inhibit the growth of calcium oxalate monohydrate (COM) and induce the formation of calcium oxalate dihydrate (COD) in varying degrees. With the decrease in Mw of GLPs, the percentage of COD increased. GLPs increased the absolute value of the Zeta potential on the crystal surface and reduced the aggregation between crystals. Cell experiments showed that the toxicity of CaOx crystal regulated by GLPs to HK-2 cells was reduced, and the cytotoxicity of CaOx crystal regulated by GLP7 with the smallest Mw was the smallest, which was consistent with the highest SOD activity, the lowest ROS and MDA levels, the lowest OPN expression level and the lowest cell necrosis rate. These results suggest that GLPs, especially GLP7, may be a potential drug for the prevention and treatment of kidney stones.
Collapse
|
6
|
Xu F, Wu Y, Yang Q, Cheng Y, Xu J, Zhang Y, Dai H, Wang B, Ma Q, Chen Y, Lin F, Wang C. Engineered Extracellular Vesicles with SHP2 High Expression Promote Mitophagy for Alzheimer's Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2207107. [PMID: 36193769 DOI: 10.1002/adma.202207107] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Mitochondrial dysfunction is a fundamental pathological feature of Alzheimer's disease (AD). However, toxicity and poor brain enrichment of existing mitophagy inducers limit their further applications. In this study, a platform for AD therapy is developed using nanosized mesenchymal-stem-cells-derived extracellular vesicles with tyrosine phosphatase-2 (SHP2) high-expression (MSC-EVs-SHP2). The high blood-brain barrier penetration ability of MSC-EVs-SHP2 is demonstrated in AD-mice, facilitating SHP2 delivery to the brain. In addition, MSC-EVs-SHP2 significantly induces mitophagy of neuronal cells, which alleviates mitochondrial damage-mediated apoptosis and NLRP3 inflammasome activation. Mitophagy further diminishes neuronal cells apoptosis and neuroinflammation, culminating with rescued synaptic loss and cognitive decline in an AD mouse model. The EV-engineering technology provides a potential platform for effective AD therapy by inducing mitophagy.
Collapse
Affiliation(s)
- Fang Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yi Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Qianyu Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Ying Cheng
- Institute of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Disease, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, 215123, P. R. China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yue Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Beilei Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yitong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Fang Lin
- Institute of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Disease, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, 215123, P. R. China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| |
Collapse
|
7
|
Sharma P, Karnam K, Mahale A, Sedmaki K, Krishna Venuganti V, Kulkarni OP. HDAC5 RNA interference ameliorates acute renal injury by upregulating KLF2 and inhibiting NALP3 expression in a mouse model of oxalate nephropathy. Int Immunopharmacol 2022; 112:109264. [DOI: 10.1016/j.intimp.2022.109264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/31/2022] [Accepted: 09/16/2022] [Indexed: 11/30/2022]
|
8
|
Xiong P, Cheng XY, Sun XY, Chen XW, Ouyang JM. Interaction between nanometer calcium oxalate and renal epithelial cells repaired with carboxymethylated polysaccharides. BIOMATERIALS ADVANCES 2022; 137:212854. [PMID: 35929244 DOI: 10.1016/j.bioadv.2022.212854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Injury of renal tubular epithelial cells (HK-2) is an important cause of kidney stone formation. In this article, the repairing effect of polysaccharide (PCP0) extracted from the traditional Chinese medicine Poria cocos and its carboxymethylated derivatives on damaged HK-2 cells was studied, and the differences in adhesion and endocytosis of the cells to nanometer calcium oxalate monohydrate (COM) before and after repair were explored. METHODS Sodium oxalate (2.8 mmol/L) was used to damage HK-2 cells to establish a damage model, and then Poria cocos polysaccharides (PCPs) with different carboxyl (COOH) contents were used to repair the damaged cells. The changes in the biochemical indicators of the cells before and after the repair and the changes in the ability to adhere to and internalize nano-COM were detected. RESULTS The natural PCPs (PCP0, COOH content = 2.56%) were carboxymethylated, and three carboxylated modified Poria cocos with 7.48% (PCP1), 12.07% (PCP2), and 17.18% (PCP3) COOH contents were obtained. PCPs could repair the damaged HK-2 cells, and the cell viability was enhanced after repair. The cell morphology was gradually repaired, the proliferation and healing rate were increased. The ROS production was reduced, and the polarity of the mitochondrial membrane potential was restored. The level of intracellular Ca2+ ions decreased, and the autophagy response was weakened. CONCLUSION The cells repaired by PCPs inhibited the adhesion to nano-COM and simultaneously promoted the endocytosis of nano-COM. The endocytic crystals mainly accumulated in the lysosome. Inhibiting adhesion and increasing endocytosis could reduce the nucleation, growth, and aggregation of cell surface crystals, thereby inhibiting the formation of kidney stones. With the increase of COOH content in PCPs, its ability to repair damaged cells, inhibit crystal adhesion, and promote crystal endocytosis all increased, that is, PCP3 with the highest COOH content showed the best ability to inhibit stone formation.
Collapse
Affiliation(s)
- Peng Xiong
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Xiao-Yan Cheng
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Xin-Yuan Sun
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Xue-Wu Chen
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
9
|
Greasley J, Goolcharan S, Andrews R. Quantitative phase analysis and microstructural characterization of urinary tract calculi with X-ray diffraction Rietveld analysis on a Caribbean island. J Appl Crystallogr 2022. [DOI: 10.1107/s1600576721011602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In the twin-island state of Trinidad and Tobago, urinary stone analysis is not routinely performed. This study investigates, via powder X-ray diffraction, 52 urinary tract calculi collected from hospitals in Trinidad. Of these, 46 stones were analysed with Rietveld refinement for quantitative analysis and materials characterization. Refined unit-cell, microstructural and weight fraction parameters were obtained, with the last being used for stone classification. The results revealed seven distinct mineralogical phases of varying frequency: calcium oxalate monohydrate (COM, 58%), calcium oxalate dihydrate (COD, 23%), carbonated apatite (APA, 48%), brushite (BRU, 6%), struvite (STR, 42%), uric acid (UA, 23%) and ammonium acid urate (AAU, 19%). The average refined crystallite sizes were 1352 ± 90 Å (COM), 1921 ± 285 Å (COD), 83 ± 5 Å (APA), 1172 ± 9 Å (BRU), 1843 ± 138 Å (STR), 981 ± 87 Å (UA) and 292 ± 83 Å (AAU). Subsequently, 36.5% of stones were categorized as phosphates, 34.6% as oxalates, 13.5% as uric acid/urates and 15.4% as mixed compositions. The study findings highlight the importance of stone analysis as a necessary step towards disease management of local patients, and endorse the application of Rietveld refinement as a natural extension to diffraction-based kidney stone investigations.
Collapse
|
10
|
Paiva WS, de Souza Neto FE, Queiroz MF, Batista LANC, Rocha HAO, de Lima Batista AC. Oligochitosan Synthesized by Cunninghamella elegans, a Fungus from Caatinga (The Brazilian Savanna) Is a Better Antioxidant than Animal Chitosan. Molecules 2021; 27:molecules27010171. [PMID: 35011403 PMCID: PMC8747077 DOI: 10.3390/molecules27010171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022] Open
Abstract
Animal chitosan (Chit-A) is gaining more acceptance in daily activities. It is used in a range of products from food supplements for weight loss to even raw materials for producing nanoparticles and hydrogel drug carriers; however, it has low antioxidant activity. Fungal oligochitosan (OChit-F) was identified as a potential substitute for Chit-A. Cunninghamella elegans is a fungus found in the Brazilian savanna (Caatinga) that produces OligoChit-F, which is a relatively poorly studied compound. In this study, 4 kDa OChit-F with a 76% deacetylation degree was extracted from C. elegans. OChit-F showed antioxidant activity similar to that of Chit-A in only one in vitro test (copper chelation) but exhibited higher activity than that of Chit-A in three other tests (reducing power, hydroxyl radical scavenging, and iron chelation). These results indicate that OChit-F is a better antioxidant than Chit-A. In addition, Chit-A significantly increased the formation of calcium oxalate crystals in vitro, particularly those of the monohydrate (COM) type; however, OChit-F had no effect on this process in vitro. In summary, OChit-F had higher antioxidant activity than Chit-A and did not induce the formation of CaOx crystals. Thus, OChit-F can be used as a Chit-A substitute in applications affected by oxidative stress.
Collapse
Affiliation(s)
- Weslley Souza Paiva
- Postgraduate Programe in Biotechnology (RENORBIO), Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Rio Grande do Norte, Brazil;
- Laboratório de Biotecnologia de Polímeros Naturais-BIOPOL, Departament of Biochemistry, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Rio Grande do Norte, Brazil; (M.F.Q.); (L.A.N.C.B.)
- Correspondence:
| | | | - Moacir Fernandes Queiroz
- Laboratório de Biotecnologia de Polímeros Naturais-BIOPOL, Departament of Biochemistry, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Rio Grande do Norte, Brazil; (M.F.Q.); (L.A.N.C.B.)
- Biomedicine Departament, Universidade Potiguar, Natal 59056-000, Rio Grande do Norte, Brazil
| | - Lucas Alighieri Neves Costa Batista
- Laboratório de Biotecnologia de Polímeros Naturais-BIOPOL, Departament of Biochemistry, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Rio Grande do Norte, Brazil; (M.F.Q.); (L.A.N.C.B.)
| | - Hugo Alexandre Oliveira Rocha
- Postgraduate Programe in Biotechnology (RENORBIO), Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, Rio Grande do Norte, Brazil;
- Biomedicine Departament, Universidade Potiguar, Natal 59056-000, Rio Grande do Norte, Brazil
| | | |
Collapse
|
11
|
Boadi EA, Shin S, Gombedza F, Bandyopadhyay BC. Differential biomolecular recognition by synthetic vs. biologically-derived components in the stone-forming process using 3D microfluidics. J Mater Chem B 2021; 10:34-46. [PMID: 34779812 PMCID: PMC9045411 DOI: 10.1039/d1tb01213d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcium phosphate (CaP) biomineralization is the hallmark of extra-skeletal tissue calcification and renal calcium stones. Although such a multistep process starts with CaP crystal formation, the mechanism is still poorly understood due to the complexity of the in vivo system and the lack of a suitable approach to simulate a truly in vivo-like environment. Although endogenous proteins and lipids are engaged with CaP crystals in such a biological process of stone formation, most in vitro studies use synthetic materials that can display differential bioreactivity and molecular recognition by the cellular component. Here, we used our in vitro microfluidic (MF) tubular structure, which is the first completely cylindrical platform, with renal tubular cellular microenvironments closest to the functional human kidney tubule, to understand the precise role of biological components in this process. We systematically evaluated the contribution of synthetic and biological components in the stone-forming process in the presence of dynamic microenvironmental cues that originated due to cellular pathophysiology, which are critical for the nucleation, aggregation, and growth of CaP crystals. Our results show that crystal aggregation and growth were enhanced by immunoglobulin G (IgG), which was further inhibited by etidronic acid due to the chelation of extracellular Ca2+. Interestingly, biogenic CaP crystals from mice urine, when applied with cell debris and non-specific protein (bovine serum albumin), exhibited a more discrete crystal growth pattern, compared to exposure to synthetic CaP crystals under similar conditions. Furthermore, proteins found on those calcium crystals from mice urine produced discriminatory effects on crystal-protein attachment. Specifically, such biogenic crystals exhibited enhanced affinity to the proteins inherent to those crystals. More importantly, a physiological comparison of crystal induction in renal tubular cells revealed that biogenic crystals are less effective at producing a sustained rise in cytosolic Ca2+ compared to synthetic crystals, suggesting a milder detrimental effect to downstream signaling. Finally, synthetic crystal-internalized cells induced more oxidative stress, inflammation, and cellular damage compared to the biogenic crystal-internalized cells. Together, these results suggest that the intrinsic nature of biogenically derived components are appropriate to generate the molecular recognition needed for spatiotemporal effects and are critical towards understanding the process of kidney stone formation.
Collapse
Affiliation(s)
- Eugenia Awuah Boadi
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA
| | - Samuel Shin
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA
| | - Farai Gombedza
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA
| | - Bidhan C. Bandyopadhyay
- Calcium Signaling Laboratory, 151 Research Service, DC Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, 20422, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington DC, 20037, USA,Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington DC, 20064, USA
| |
Collapse
|
12
|
Singh A, Tandon S, Nandi SP, Kaur T, Tandon C. Downregulation of inflammatory mediators by ethanolic extract of Bergenia ligulata (Wall.) in oxalate injured renal epithelial cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114104. [PMID: 33836258 DOI: 10.1016/j.jep.2021.114104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/05/2021] [Accepted: 04/03/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the Indian traditional system of medicine, Bergenia ligulata (Wall.) Engl. has been used for treatment of urolithiasis. Its efficacious nature has led to its incorporation in various commercial herbal formulations such as Cystone and Neeri which are prescribed for kidney related ailments. AIM OF THE STUDY To assess whether ethanolic extract of B. ligulata can mitigate the cascade of inflammatory responses that cause oxidative stress and ultimately cell death in renal epithelial cells exposed to hyperoxaluric conditions. MATERIAL AND METHODS Bioactivity guided fractionation using solvents of varying polarities was employed to evaluate the potential of the extracts of B. ligulata to inhibit the crystallization process. Modulation of crystal morphology was visualized through Scanning electron microscopy (SEM) analysis. Cell death was assessed using flow cytometry based assays. Alteration in the inflammatory mediators was evaluated using real time PCR and immunocytochemistry. Phytochemical characterization of the ethanolic extract was carried out using FTIR, LC-MS and GC-MS. RESULTS Bioactivity guided fractionation for the assessment of antilithiatic activity revealed dose dependent inhibition of nucleation and aggregation process of calcium oxalate crystals in the presence of various extracts, however ethanolic extract showed maximum inhibition and was chosen for further experiments. Studies on renal epithelial NRK-52E cells showed, cytoprotective efficacy of B. ligulata extract against oxalate injury. SEM anaysis further revealed the potential of the extract to modulate the crystal structure and adhesion to renal cell surface. Exposure of the renal cells to the extract led to conversion of the calcium oxalate monohydrate (COM) crystals to the less injurious calcium oxalate dihydrate (COD) form. Expression analysis for oxidative stress and inflammatory biomarkers in NRK-52E cells revealed up-regulation of Mitogen activated protein kinase (MAPK), Osteopontin (OPN) and Nuclear factor- ĸB (NF-ĸB), in response to calcium oxalate insult; which was drastically reduced in the presence of B. ligulata extract. Flow cytometric evaluation pointed to caspase 3 mediated apoptotic cell death in oxalate injured cells, which was attenuated by B. ligulata extract. CONCLUSION Considering the complex multifactorial etiology of urolithiasis, ethanolic extract from B. ligulata can be a promising option for the management of kidney stones, as it has the potential to limit inflammation and the subsequent cell death.
Collapse
Affiliation(s)
- Anubha Singh
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Shoma Paul Nandi
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | | |
Collapse
|
13
|
Zhang H, Sun XY, Chen XW, Ouyang JM. Degraded Porphyra yezoensis polysaccharide protects HK-2 cells and reduces nano-COM crystal toxicity, adhesion and endocytosis. J Mater Chem B 2021; 8:7233-7252. [PMID: 32638810 DOI: 10.1039/d0tb00360c] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We studied the protection of degraded Porphyra yezoensis polysaccharide (PYP) on human proximal tubular epithelial cells (HK-2) from cytotoxicity of nano-calcium oxalate monohydrate (COM) crystal, and the regulation of adhesion and endocytosis of the COM crystal. Four degraded fractions, namely, PYP1, PYP2, PYP3, and PYP4, were successfully obtained, with molecular weights (Mws) of 576.2, 49.5, 12.6, and 4.02 kDa, respectively. PYP protection reduced the crystal toxicity, prevented the destruction of cell morphology and cytoskeleton, inhibited the production of reactive oxygen species and the decline of lysosomal integrity, and reduced the expression of osteopontin and transmembrane protein (CD44). PYPi inhibited the adhesion and endocytosis of HK-2 cells by nano-COM. Endocytic COM crystals were accumulated in the lysosomes. With decreasing molecular weight, the ability of PYP to reduce cell damage and inhibit cell adhesion and endocytosis increased. PYP4, which has the smallest molecular weight, weaker intramolecular hydrogen bonds and more reducing groups, showed the best biological activity. PYPi can reduce the oxidative damage of the crystal to the cell, inhibit the adhesion and endocytosis of the crystal, and reduce the risk of kidney stone formation. Therefore, PYP, especially PYP4, has potential for use as a green drug to inhibit the formation and recurrence of calcium oxalate stones.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Chemistry, Jinan University, Guangzhou 510632, China. and Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Xin-Yuan Sun
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Xue-Wu Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China. and Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Department of Chemistry, Jinan University, Guangzhou 510632, China. and Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou 510632, China
| |
Collapse
|
14
|
Arizmendi-Izazaga A, Navarro-Tito N, Jiménez-Wences H, Mendoza-Catalán MA, Martínez-Carrillo DN, Zacapala-Gómez AE, Olea-Flores M, Dircio-Maldonado R, Torres-Rojas FI, Soto-Flores DG, Illades-Aguiar B, Ortiz-Ortiz J. Metabolic Reprogramming in Cancer: Role of HPV 16 Variants. Pathogens 2021; 10:pathogens10030347. [PMID: 33809480 PMCID: PMC7999907 DOI: 10.3390/pathogens10030347] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/13/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic reprogramming is considered one of the hallmarks in cancer and is characterized by increased glycolysis and lactate production, even in the presence of oxygen, which leads the cancer cells to a process called “aerobic glycolysis” or “Warburg effect”. The E6 and E7 oncoproteins of human papillomavirus 16 (HPV 16) favor the Warburg effect through their interaction with a molecule that regulates cellular metabolism, such as p53, retinoblastoma protein (pRb), c-Myc, and hypoxia inducible factor 1α (HIF-1α). Besides, the impact of the E6 and E7 variants of HPV 16 on metabolic reprogramming through proteins such as HIF-1α may be related to their oncogenicity by favoring cellular metabolism modifications to satisfy the energy demands necessary for viral persistence and cancer development. This review will discuss the role of HPV 16 E6 and E7 variants in metabolic reprogramming and their contribution to developing and preserving the malignant phenotype of cancers associated with HPV 16 infection.
Collapse
Affiliation(s)
- Adán Arizmendi-Izazaga
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (N.N.-T.); (M.O.-F.)
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Miguel A. Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
| | - Dinorah N. Martínez-Carrillo
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Ana E. Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (N.N.-T.); (M.O.-F.)
| | - Roberto Dircio-Maldonado
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
- Laboratorio de Diagnóstico e Investigación en Salud, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Francisco I. Torres-Rojas
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Diana G. Soto-Flores
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
- Laboratorio de Diagnóstico e Investigación en Salud, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
- Correspondence: ; Tel.: +52-747-471-0901
| |
Collapse
|
15
|
Sun XY, Zhang H, Chen JY, Zeng GH, Ouyang JM. Porphyra yezoensis polysaccharide and potassium citrate synergistically inhibit calcium oxalate crystallization induced by renal epithelial cells and cytotoxicity of the formed crystals. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 119:111448. [DOI: 10.1016/j.msec.2020.111448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/19/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
|
16
|
Differential bound proteins and adhesive capabilities of calcium oxalate monohydrate crystals with various sizes. Int J Biol Macromol 2020; 163:2210-2223. [PMID: 32956748 DOI: 10.1016/j.ijbiomac.2020.09.085] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/02/2020] [Accepted: 09/12/2020] [Indexed: 12/22/2022]
Abstract
Adhesion of calcium oxalate (CaOx) crystals onto renal tubular epithelial cells is one of the critical steps in kidney stone formation. However, effects of crystal size on the crystal adhesive capability remained unclear. This study compared the adhesive capabilities of CaOx monohydrate (COM) crystals with various sizes (<10 μm, 20-30 μm, 50-60 μm, and > 80 μm). Crystal-cell adhesion assay showed size-dependent increase of COM crystal adhesion onto epithelial cell surface using the larger crystals. Identification of apical membrane proteins that could bind to COM crystals by tandem mass spectrometry (nanoLC-ESI-ETD MS/MS) demonstrated size-specific sets of the COM crystal-binding proteins. Among these, numbers of known oxalate-binding proteins and COM crystal receptors were greatest in the set of the largest size (>80 μm). Atomic force microscopy (AFM) revealed that adhesive forces between carboxylic-immobilized AFM tip and COM crystal surface and between COM-mounted AFM tip and renal epithelial cell surface were size-dependent (greater for the larger crystals). In summary, the adhesive capability of COM crystals is size-dependent - the larger the greater adhesive capability. These data may help better understanding of the pathogenic mechanisms of kidney stone formation at an initial stage when renal tubular cells are exposed to various sizes of COM crystals.
Collapse
|
17
|
Modi PK, Prabhu A, Bhandary YP, Shenoy P. S, Hegde A, ES SP, Johnson RP, Das SP, Vazirally S, Rekha PD. Effect of calcium glucoheptonate on proliferation and osteogenesis of osteoblast-like cells in vitro. PLoS One 2019; 14:e0222240. [PMID: 31498830 PMCID: PMC6733474 DOI: 10.1371/journal.pone.0222240] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Calcium is the key macromineral having a role in skeletal structure and function, muscle contraction, and neurotransmission. Bone remodeling is maintained through a constant balance between calcium resorption and deposition. Calcium deficiency is resolved through calcium supplementation, and among the supplements, water-soluble organic molecules attracted great pharmaceutical interest. Calcium glucoheptonate is a highly water-soluble organic calcium salt having clinical use; however, detailed investigations on its biological effects are limited. We assessed the effects of calcium glucoheptonate on cell viability and proliferation of osteoblast-like MG-63 cells. Calcium uptake and mineralization were evaluated using Alizarin red staining of osteoblast-like MG-63 cells treated with calcium glucoheptonate. Expression of osteogenic markers were monitored by western blotting, immunofluorescence, and qRT-PCR assays. Increased proliferation and calcium uptake were observed in the MG-63 cells treated with calcium glucoheptonate. The treatment also increased the expression of osteopontin and osteogenic genes such as collagen-1, secreted protein acidic and cysteine rich (SPARC), and osteocalcin. Calcium glucoheptonate treatment did not exert any cytotoxicity on colorectal and renal epithelial cells, indicating the safety of the treatment. This is the first report with evidence for its beneficial effect for pharmaceutical use in addressing calcium deficiency conditions.
Collapse
Affiliation(s)
- Prashant Kumar Modi
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Ashwini Prabhu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Yashodhar P. Bhandary
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sudheer Shenoy P.
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Aparna Hegde
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sindhu Priya ES
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Renjith P. Johnson
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Shankar Prasad Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | | | | |
Collapse
|
18
|
Gombedza FC, Shin S, Kanaras YL, Bandyopadhyay BC. Abrogation of store-operated Ca 2+ entry protects against crystal-induced ER stress in human proximal tubular cells. Cell Death Discov 2019; 5:124. [PMID: 31396401 PMCID: PMC6680047 DOI: 10.1038/s41420-019-0203-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/24/2019] [Accepted: 07/07/2019] [Indexed: 12/15/2022] Open
Abstract
Calcium crystal internalization into proximal tubular (PT) cells results in acute kidney injury, nephrocalcinosis, chronic kidney disease (CKD), and kidney-stone formation. Ca2+ supersaturation in PT luminal fluid induces calcium crystal formation, leading to aberrant crystal internalization into PT cells. While such crystal internalization produces reactive oxygen species (ROS), cell membrane damage, and apoptosis; the upstream signaling events involving dysregulation of intracellular Ca2+ homeostasis and ER stress, remain largely unknown. We have recently described a transepithelial Ca2+ transport pathway regulated by receptor-operated Ca2+ entry (ROCE) in PT cells. Therefore, we examined the pathophysiological consequence of internalization of stone-forming calcium crystals such as calcium phosphate (CaP), calcium oxalate (CaOx), and CaP + CaOx (mixed) crystals on the regulation of intracellular Ca2+ signaling by measuring dynamic changes in Ca2+ transients in HK2, human PT cells, using pharmacological and siRNA inhibitors. The subsequent effect on ER stress was measured by changes in ER morphology, ER stress-related gene expression, endogenous ROS production, apoptosis, and necrosis. Interestingly, our data show that crystal internalization induced G-protein-coupled receptor-mediated sustained rise in intracellular Ca2+ concentration ([Ca2+]i) via store-operated Ca2+ entry (SOCE); suggesting that the mode of Ca2+ entry switches from ROCE to SOCE following crystal internalization. We found that SOCE components-stromal interacting molecules 1 and 2 (STIM1, STIM2) and ORAI3 (SOCE) channel were upregulated in these crystal-internalized cells, which induced ER stress, ROS production, and cell death. Finally, silencing those SOCE genes protected crystal-internalized cells from prolonged [Ca2+]i rise and ER stress. Our data provide insight into the molecular mechanism of crystal-induced Ca2+ dysregulation, ER stress, and PT cell death and thus could have a translational role in treating crystal nephropathies including kidney stones. Taken together, modulation of Ca2+ signaling can be used as a tool to reverse the pathological consequence of crystal-induced conditions including cardiovascular calcification.
Collapse
Affiliation(s)
- Farai C. Gombedza
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street NW, Washington, DC 20422 USA
| | - Samuel Shin
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street NW, Washington, DC 20422 USA
| | - Yianni L. Kanaras
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street NW, Washington, DC 20422 USA
| | - Bidhan C. Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street NW, Washington, DC 20422 USA
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, 2150 Pennsylvania Avenue NW, Washington, DC 20037 USA
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064 USA
| |
Collapse
|
19
|
Priante G, Gianesello L, Ceol M, Del Prete D, Anglani F. Cell Death in the Kidney. Int J Mol Sci 2019; 20:E3598. [PMID: 31340541 PMCID: PMC6679187 DOI: 10.3390/ijms20143598] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Apoptotic cell death is usually a response to the cell's microenvironment. In the kidney, apoptosis contributes to parenchymal cell loss in the course of acute and chronic renal injury, but does not trigger an inflammatory response. What distinguishes necrosis from apoptosis is the rupture of the plasma membrane, so necrotic cell death is accompanied by the release of unprocessed intracellular content, including cellular organelles, which are highly immunogenic proteins. The relative contribution of apoptosis and necrosis to injury varies, depending on the severity of the insult. Regulated cell death may result from immunologically silent apoptosis or from immunogenic necrosis. Recent advances have enhanced the most revolutionary concept of regulated necrosis. Several modalities of regulated necrosis have been described, such as necroptosis, ferroptosis, pyroptosis, and mitochondrial permeability transition-dependent regulated necrosis. We review the different modalities of apoptosis, necrosis, and regulated necrosis in kidney injury, focusing particularly on evidence implicating cell death in ectopic renal calcification. We also review the evidence for the role of cell death in kidney injury, which may pave the way for new therapeutic opportunities.
Collapse
Affiliation(s)
- Giovanna Priante
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy.
| | - Lisa Gianesello
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Monica Ceol
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Dorella Del Prete
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Franca Anglani
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
20
|
Zhang H, Sun XY, Ouyang JM. Effects of Porphyra yezoensis Polysaccharide with Different Molecular Weights on the Adhesion and Endocytosis of Nanocalcium Oxalate Monohydrate in Repairing Damaged HK-2 Cells. ACS Biomater Sci Eng 2019; 5:3974-3986. [PMID: 33443420 DOI: 10.1021/acsbiomaterials.9b00410] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Hui Zhang
- Institute of Biomineralization and Lithiasis Research, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Xin-Yuan Sun
- Institute of Biomineralization and Lithiasis Research, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| |
Collapse
|
21
|
Cell death in ectopic calcification of the kidney. Cell Death Dis 2019; 10:466. [PMID: 31197147 PMCID: PMC6565680 DOI: 10.1038/s41419-019-1697-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 05/17/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022]
|
22
|
Gallic Acid-Chitosan Conjugate Inhibits the Formation of Calcium Oxalate Crystals. Molecules 2019; 24:molecules24112074. [PMID: 31151328 PMCID: PMC6600518 DOI: 10.3390/molecules24112074] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/19/2019] [Accepted: 05/22/2019] [Indexed: 11/23/2022] Open
Abstract
It has recently been shown that chitosan (Chit) induces the formation of calcium oxalate (CaOx) crystals, which are mainly responsible for the appearance of kidney stones, and this might limit the use of Chit in vivo. Here, Chit was conjugated with gallic acid (Chit-Gal) to decrease the formation of CaOx crystal. This conjugation was confirmed by FTIR and NMR analyses. Chit-Gal contains 10.2 ± 1.5 mg GA per g of Chit. Compared to the control group, Chit increased the number of crystals by six-fold, mainly in the number of monohydrated CaOx crystals, which are the most harmful CaOx crystals. In addition, Chit increased the zeta potential (ζ) of CaOx crystals by three-fold, indicating that Chit was associated with the crystals. These alterations were abolished when Chit-gal was used in these tests. As oxidative stress is related to renal calculus formation, Chit and Chit-Gal were also evaluated as antioxidants using total antioxidant Capacity (TAC), reducing power, ferrous chelation, and copper chelation tests. Chit-gal was more efficient antioxidant agent in TAC (2 times), in ferrous chelation (90 times), and in reducing Power (5 times) than Chit. Overall, Chit-gal has higher antioxidant activity than Chit, does not induce the formation of CaOx crystals. Thus, Chit-Gal has potential to be used as a chit substitute.
Collapse
|
23
|
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: 3.0] [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
|
24
|
Sun XY, Zhang H, Liu J, Ouyang JM. Repair activity and crystal adhesion inhibition of polysaccharides with different molecular weights from red algae Porphyra yezoensis against oxalate-induced oxidative damage in renal epithelial cells. Food Funct 2019; 10:3851-3867. [DOI: 10.1039/c8fo02556h] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Porphyra yezoensis polysaccharide repaired oxalate-injured renal epithelial cells and decreased COM crystal adhesion on the cell surface.
Collapse
Affiliation(s)
- Xin-Yuan Sun
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
- Institute of Biomineralization and Lithiasis Research
| | - Hui Zhang
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
- Institute of Biomineralization and Lithiasis Research
| | - Jie Liu
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Jian-Ming Ouyang
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
- Institute of Biomineralization and Lithiasis Research
| |
Collapse
|
25
|
Wilson GJ, Gois PHF, Zhang A, Wang X, Law BMP, Kassianos AJ, Healy HG. The Role of Oxidative Stress and Inflammation in Acute Oxalate Nephropathy Associated With Ethylene Glycol Intoxication. Kidney Int Rep 2018; 3:1217-1221. [PMID: 30197989 PMCID: PMC6127410 DOI: 10.1016/j.ekir.2018.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Gregory J Wilson
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,School of Clinical Medicine, Royal Brisbane Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Pedro H F Gois
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,School of Clinical Medicine, Royal Brisbane Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Alice Zhang
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,School of Clinical Medicine, Royal Brisbane Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia
| | - Xiangju Wang
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia
| | - Becker M P Law
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Andrew J Kassianos
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,School of Clinical Medicine, Royal Brisbane Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Helen G Healy
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,School of Clinical Medicine, Royal Brisbane Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
26
|
Sun XY, Ouyang JM, Bhadja P, Gui Q, Peng H, Liu J. Protective Effects of Degraded Soybean Polysaccharides on Renal Epithelial Cells Exposed to Oxidative Damage. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7911-7920. [PMID: 27701856 DOI: 10.1021/acs.jafc.6b03323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
This study aimed to investigate the protective effects of degraded soybean polysaccharides (DSP) on oxidatively damaged African green monkey kidney epithelial (Vero) cells. Low DSP concentration (10 μg/mL) elicited an evident protective effect on H2O2-induced cell injury (0.3 mmol/L). The cell viabilities of the H2O2-treated group and the DSP-protected group were 57.3 and 93.1%, respectively. The cell viability decreased to 88.3% when the dosage was increased to 100 μg/mL. DSP protected Vero cells from H2O2-mediated oxidative damage by enhancing cellular superoxide dismutase activity and total antioxidant capacity and by decreasing malonaldehyde content and lactate dehydrogenase release. The H2O2-treated cells stimulated the aggregation of calcium oxalate monohydrate crystals. DSP could also reduce the crystal size, decrease the attached crystal content, and prevent the cell aggregation by alleviating oxidative injury and lipid peroxidation, enhancing antioxidant capacity, and decreasing hyaluronan expression on cellular surfaces. The internalization ability of the injured cells was improved after these cells were exposed to DSP solution. The regulation ability of DSP-repaired cells on calcium oxalate dihydrate formation, crystal attachment, aggregation, and internalization was lower than that of normal cells but was higher than that of the injured cells. DSP may be a potential green drug to prevent calcium oxalate (CaOx) stone formation because DSP could protect cells from oxidative damage and inhibit CaOx crystal formation.
Collapse
Affiliation(s)
- Xin-Yuan Sun
- Department of Chemistry and ‡Institute of Biomineralization and Lithiasis Research, Jinan University , Guangzhou 510632, China
| | - Jian-Ming Ouyang
- Department of Chemistry and ‡Institute of Biomineralization and Lithiasis Research, Jinan University , Guangzhou 510632, China
| | - Poonam Bhadja
- Department of Chemistry and ‡Institute of Biomineralization and Lithiasis Research, Jinan University , Guangzhou 510632, China
| | - Qin Gui
- Department of Chemistry and ‡Institute of Biomineralization and Lithiasis Research, Jinan University , Guangzhou 510632, China
| | - Hua Peng
- Department of Chemistry and ‡Institute of Biomineralization and Lithiasis Research, Jinan University , Guangzhou 510632, China
| | - Jie Liu
- Department of Chemistry and ‡Institute of Biomineralization and Lithiasis Research, Jinan University , Guangzhou 510632, China
| |
Collapse
|
27
|
Affiliation(s)
- X-Y Sun
- Department of Chemistry, Jinan University, Guangzhou, China
| | - J-M Ouyang
- Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|