Доступ предоставлен для: Guest
Critical Reviews™ in Eukaryotic Gene Expression

Выходит 6 номеров в год

ISSN Печать: 1045-4403

ISSN Онлайн: 2162-6502

The Impact Factor measures the average number of citations received in a particular year by papers published in the journal during the two preceding years. 2017 Journal Citation Reports (Clarivate Analytics, 2018) IF: 1.6 To calculate the five year Impact Factor, citations are counted in 2017 to the previous five years and divided by the source items published in the previous five years. 2017 Journal Citation Reports (Clarivate Analytics, 2018) 5-Year IF: 2.2 The Immediacy Index is the average number of times an article is cited in the year it is published. The journal Immediacy Index indicates how quickly articles in a journal are cited. Immediacy Index: 0.3 The Eigenfactor score, developed by Jevin West and Carl Bergstrom at the University of Washington, is a rating of the total importance of a scientific journal. Journals are rated according to the number of incoming citations, with citations from highly ranked journals weighted to make a larger contribution to the eigenfactor than those from poorly ranked journals. Eigenfactor: 0.00058 The Journal Citation Indicator (JCI) is a single measurement of the field-normalized citation impact of journals in the Web of Science Core Collection across disciplines. The key words here are that the metric is normalized and cross-disciplinary. JCI: 0.33 SJR: 0.345 SNIP: 0.46 CiteScore™:: 2.5 H-Index: 67

Indexed in

Mechanisms Mediating Oxalate-Induced Alterations in Renal Cell Functions

Том 13, Выпуск 1, 2003, 18 pages
DOI: 10.1615/CritRevEukaryotGeneExpr.v13.i1.50
Get accessGet access

Краткое описание

Oxalate is a major component of the most common form of kidney stones—calcium oxalate stones. High concentrations of oxalate promote stone formation in two ways: (1) by providing urinary conditions favorable to the formation of calcium oxalate crystals, and (2) by inducing renal injury that generates cellular debris and promotes crystal nucleation and attachment. Oxalate toxicity is mediated in part by activation of lipid signaling pathways that produce arachidonic acid, lysophospholipids, and ceramide. These lipids disrupt mitochondrial function by increasing reactive oxygen species (ROS), decreasing mitochondrial membrane potential, and increasing mitochondrial permeability. The net response is cytochrome C release, activation of caspases, and apoptosis or necrosis. Not all cells succumb to oxalate toxicity, however; in those cells that don’t, ROS and lipid-signaling molecules induce changes in gene expression that allow them to survive and adapt to the toxic insult. The increased expression of immediate early genes (IEGs), osteopontin, extracellular matrix (ECM) proteins, crystallization inhibitors, and chemokines orchestrates a group of cellular responses—including cell proliferation, secretion of kidney stone inhibitory proteins, recruitment of immune cells, and tissue remodeling—that limit accumulation of cell debris or increase the production of inhibitors of calcium oxalate crystallization, thereby limiting stone formation.

ЦИТИРОВАНО В
  1. Escobar Carla, Byer Karen J., Khan Saeed R., Naturally produced crystals obtained from kidney stones are less injurious to renal tubular epithelial cells than synthetic crystals, BJU International, 100, 4, 2007. Crossref

  2. UMEKAWA TOHRU, IGUCHI MASANORI, UEMURA HIROTSUGU, KHAN SAEED R., Oxalate ions and calcium oxalate crystal-induced up-regulation of osteopontin and monocyte chemoattractant protein-1 in renal fibroblasts, BJU International, 98, 3, 2006. Crossref

  3. Baran Enrique J., Monje Paula V., Oxalate Biominerals, in Biomineralization, 2010. Crossref

  4. Steenkamp Vanessa, Stewart Michael J, Nephrotoxicity Associated With Exposure to Plant Toxins, With Particular Reference to Africa, Therapeutic Drug Monitoring, 27, 3, 2005. Crossref

  5. Grover PK, Miyazawa K, Coleman M, Stahl J, Ryall RL, Renal prothrombin mRNA is significantly decreased in a hyperoxaluric rat model of nephrolithiasis, The Journal of Pathology, 210, 3, 2006. Crossref

  6. Jonassen Julie A., Kohjimoto Yasuo, Scheid Cheryl R., Schmidt Madelyn, Oxalate toxicity in renal cells, Urological Research, 33, 5, 2005. Crossref

  7. Khaskhali Muhammad H., Byer Karen J., Khan Saeed R., The effect of calcium on calcium oxalate monohydrate crystal-induced renal epithelial injury, Urological Research, 37, 1, 2009. Crossref

  8. Holmes Ross P., Assimos Dean G., The impact of dietary oxalate on kidney stone formation, Urological Research, 32, 5, 2004. Crossref

  9. Marengo Susan Ruth, Chen Daniel H. -C., Evan Andrew P., Sommer Andre J., Stowe Nicholas T., Ferguson Donald G., Resnick Martin I., MacLennan Gregory T., Continuous infusion of oxalate by minipumps induces calcium oxalate nephrocalcinosis, Urological Research, 34, 3, 2006. Crossref

  10. Guo Xiaomei, Stotz Henrik U., ABA signaling inhibits oxalate-induced production of reactive oxygen species and protects against Sclerotinia sclerotiorum in Arabidopsis thaliana, European Journal of Plant Pathology, 128, 1, 2010. Crossref

  11. Khan Saeed R., Hyperoxaluria-induced oxidative stress and antioxidants for renal protection, Urological Research, 33, 5, 2005. Crossref

  12. Marengo Susan R, Romani Andrea MP, Oxalate in renal stone disease: the terminal metabolite that just won't go away, Nature Clinical Practice Nephrology, 4, 7, 2008. Crossref

  13. Chen Shushang, Gao Xiaofeng, Sun Yinghao, Xu Chuanliang, Wang Linhui, Zhou Tie, Analysis of HK-2 cells exposed to oxalate and calcium oxalate crystals: proteomic insights into the molecular mechanisms of renal injury and stone formation, Urological Research, 38, 1, 2010. Crossref

  14. Umekawa Tohru, Tsuji Hidenori, Uemura Hirotsugu, Khan Saeed R., Superoxide from NADPH oxidase as second messenger for the expression of osteopontin and monocyte chemoattractant protein-1 in renal epithelial cells exposed to calcium oxalate crystals, BJU International, 104, 1, 2009. Crossref

  15. HOLMES ROSS P., AMBROSIUS WALTER T., ASSIMOS DEAN G., DIETARY OXALATE LOADS AND RENAL OXALATE HANDLING, Journal of Urology, 174, 3, 2005. Crossref

  16. Canales Benjamin K., Anderson Lorraine, Higgins Leeann, Slaton Joel, Roberts Ken P., Liu Nathan, Monga Manoj, Second Prize:Comprehensive Proteomic Analysis of Human Calcium Oxalate Monohydrate Kidney Stone Matrix, Journal of Endourology, 22, 6, 2008. Crossref

  17. Reply by Authors, Journal of Urology, 175, 4, 2006. Crossref

  18. Fang Hua-Chang, Lee Po-Tsang, Lu Pei-Jung, Chen Chien-Liang, Chang Tsu-Yuan, Hsu Chih-Yang, Chung Hsiao-Min, Chou Kang-Ju, Mechanisms of star fruit-induced acute renal failure, Food and Chemical Toxicology, 46, 5, 2008. Crossref

  19. Veena Coothan Kandaswamy, Josephine Anthony, Preetha Sreenivasan P., Rajesh Nachiappa Ganesh, Varalakshmi Palaninathan, Mitochondrial dysfunction in an animal model of hyperoxaluria: A prophylactic approach with fucoidan, European Journal of Pharmacology, 579, 1-3, 2008. Crossref

  20. Escobar Carla, Byer Karen J., Khaskheli Hassan, Khan Saeed R., Apatite Induced Renal Epithelial Injury: Insight Into the Pathogenesis of Kidney Stones, Journal of Urology, 180, 1, 2008. Crossref

  21. Rajeswari Arjunan, Varalakshmi Palaninathan, Low molecular weight heparin protection against oxalate-induced oxidative renal insult, Clinica Chimica Acta, 370, 1-2, 2006. Crossref

  22. Khan Saeed R., Is oxidative stress, a link between nephrolithiasis and obesity, hypertension, diabetes, chronic kidney disease, metabolic syndrome?, Urological Research, 40, 2, 2012. Crossref

  23. Khan Aslam, Byer Karen, Khan Saeed R., Exposure of Madin-Darby Canine Kidney (MDCK) Cells to Oxalate and Calcium Oxalate Crystals Activates Nicotinamide Adenine Dinucleotide Phosphate (NADPH)-Oxidase, Urology, 83, 2, 2014. Crossref

  24. Saha Sarmistha, Goswami Gagan, Pandrangi Anupama, Isolation and prevention of calcium oxalate-induced apoptotic death and oxidative stress in MDCK cells by diosgenin, Chemico-Biological Interactions, 224, 2014. Crossref

  25. Hu Yang-Yang, Dong Wei-Da, Xu Yun-Fei, Yao Xu-Dong, Peng Bo, Liu Min, Zheng Jun-Hua, Elevated Levels of miR-155 in Blood and Urine from Patients with Nephrolithiasis, BioMed Research International, 2014, 2014. Crossref

  26. Wang Shuo, Du Peng, Zhang Ning, Liu Jia, Tang Xingxing, Zhao Qiang, Yang Yong, Oligomeric proanthocyanidins protect against HK-2 cell injury induced by oxalate and calcium oxalate monohydrate crystals, Urolithiasis, 44, 3, 2016. Crossref

  27. Ligon Colin B., Hummers Laura K., McMahan Zsuzsanna H., Oxalate nephropathy in systemic sclerosis: Case series and review of the literature, Seminars in Arthritis and Rheumatism, 45, 3, 2015. Crossref

  28. Aggarwal Deepika, Gautam Diksha, Sharma Minu, Singla S.K., Bergenin attenuates renal injury by reversing mitochondrial dysfunction in ethylene glycol induced hyperoxaluric rat model, European Journal of Pharmacology, 791, 2016. Crossref

  29. Mittal Amisha, Tandon Simran, Singla Surender Kumar, Tandon Chanderdeep, Sen Utpal, Mechanistic Insights into the Antilithiatic Proteins from Terminalia arjuna: A Proteomic Approach in Urolithiasis, PLOS ONE, 11, 9, 2016. Crossref

  30. Suryavanshi Mangesh V., Bhute Shrikant S., Jadhav Swapnil D., Bhatia Manish S., Gune Rahul P., Shouche Yogesh S., Hyperoxaluria leads to dysbiosis and drives selective enrichment of oxalate metabolizing bacterial species in recurrent kidney stone endures, Scientific Reports, 6, 1, 2016. Crossref

  31. Sohgaura Atul, Bigoniya Papiya, A Review on Epidemiology and Etiology of Renal Stone, American Journal of Drug Discovery and Development, 7, 2, 2017. Crossref

  32. Meimaridou Eirini, Lobos Edgar, Hothersall John S., Renal oxidative vulnerability due to changes in mitochondrial-glutathione and energy homeostasis in a rat model of calcium oxalate urolithiasis, American Journal of Physiology-Renal Physiology, 291, 4, 2006. Crossref

  33. Rodgers Allen L., Jappie-Mahomed Dalielah, van Jaarsveld Paul J., Different effects of γ-linolenic acid (GLA) supplementation on plasma and red blood cell phospholipid fatty acid composition and calcium oxalate kidney stone risk factors in healthy subjects from two race groups with different risk profiles pose questions about the GLA-arachidonic acid-oxaluria metabolic pathway: pilot study, Urolithiasis, 46, 2, 2018. Crossref

  34. Sujatha D., Singh Kiranpal, Vohra Mursalin, Kumar K. Vijay, Sunitha S., Antilithiatic Activity of phlorotannin rich extract of <italic>Sarghassum Wightii</italic> on Calcium Oxalate Urolithiais – <italic>In Vitro</italic> and <italic>In Vivo</italic>Evaluation, International braz j urol, 41, 3, 2015. Crossref

  35. Genuis Stephen J., Kyrillos Edmond, The chemical disruption of human metabolism, Toxicology Mechanisms and Methods, 27, 7, 2017. Crossref

  36. Han Ho Jae, Lim Min Jin, Lee Yun Jung, Oxalate inhibits renal proximal tubule cell proliferation via oxidative stress, p38 MAPK/JNK, and cPLA2 signaling pathways, American Journal of Physiology-Cell Physiology, 287, 4, 2004. Crossref

  37. Jiang Hongyang, Gao Xintao, Gong Jianan, Yang Qian, Lan Ruzhu, Wang Tao, Liu Jihong, Yin Chunping, Wang Shaogang, Liu Zhuo, Downregulated Expression of Solute Carrier Family 26 Member 6 in NRK-52E Cells Attenuates Oxalate-Induced Intracellular Oxidative Stress, Oxidative Medicine and Cellular Longevity, 2018, 2018. Crossref

  38. Mittal Amisha, Tandon Simran, Singla Surender Kumar, Tandon Chanderdeep, Modulation of lithiatic injury to renal epithelial cells by aqueous extract of Terminalia arjuna, Journal of Herbal Medicine, 13, 2018. Crossref

  39. Jiang Hongyang, Pokhrel Gaurab, Chen Yinwei, Wang Tao, Yin Chunping, Liu Jihong, Wang Shaogang, Liu Zhuo, High expression of SLC26A6 in the kidney may contribute to renal calcification via an SLC26A6-dependent mechanism, PeerJ, 6, 2018. Crossref

  40. Yousefi Ghale-Salimi Mahboubeh, Eidi Maryam, Ghaemi Nasser, Khavari-Nejad Ramezan Ali, Antiurolithiatic effect of the taraxasterol on ethylene glycol induced kidney calculi in male rats, Urolithiasis, 46, 5, 2018. Crossref

  41. Sharma Minu, Naura Amarjit S., Singla S. K., Modulatory effect of 4-phenyl butyric acid on hyperoxaluria-induced renal injury and inflammation, Molecular and Cellular Biochemistry, 451, 1-2, 2019. Crossref

  42. Peerapen Paleerath, Chaiyarit Sakdithep, Thongboonkerd Visith, Protein Network Analysis and Functional Studies of Calcium Oxalate Crystal-Induced Cytotoxicity in Renal Tubular Epithelial Cells, PROTEOMICS, 18, 8, 2018. Crossref

  43. Verhulst Anja, De Broe Marc E., Oxalate, in Clinical Nephrotoxins, 2008. Crossref

  44. Gueler Faikah, Rong Song, Mengel Michael, Park Joon-Keun, Kiyan Julia, Kirsch Torsten, Dumler Inna, Haller Hermann, Shushakova Nelli, Renal Urokinase-Type Plasminogen Activator (uPA) Receptor but not uPA Deficiency Strongly Attenuates Ischemia Reperfusion Injury and Acute Kidney Allograft Rejection, The Journal of Immunology, 181, 2, 2008. Crossref

  45. Khan Saeed R., Renal Cellular Dysfunction/Damage and the Formation of Kidney Stones, in Urinary Tract Stone Disease, 2010. Crossref

  46. Wang Zhenghao, Bai Yunjin, Wang Jiahao, Wang Jia, The preventive and therapeutic effects of α-lipoic acid on ethylene glycol-induced calcium oxalate deposition in rats, International Urology and Nephrology, 52, 7, 2020. Crossref

  47. Meng Caifeng, Liu Ruina, Wang Wei, Guo Wei, Ma Hongyun, Xie Shuang, Liu Yimeng, Wang Chunying, Metabolic profiling comparison of isovitexin in normal and kidney stone model rats by ultra‐high‐performance liquid chromatography coupled to quadrupole time‐of‐flight mass spectrometry, Journal of Separation Science, 43, 12, 2020. Crossref

  48. Chaiyarit Sakdithep, Thongboonkerd Visith, Mitochondrial Dysfunction and Kidney Stone Disease, Frontiers in Physiology, 11, 2020. Crossref

  49. Wigner Paulina, Grębowski Radosław, Bijak Michal, Szemraj Janusz, Saluk-Bijak Joanna, The Molecular Aspect of Nephrolithiasis Development, Cells, 10, 8, 2021. Crossref

  50. Chen Wei, Liu Wen-Rui, Hou Jie-Bin, Ding Jia-Rong, Peng Zhong-Jiang, Gao Song-Yan, Dong Xin, Ma Jun-Hua, Lin Qi-Shan, Lu Jian-Rao, Guo Zhi-Yong, Metabolomic analysis reveals a protective effect of Fu-Fang-Jin-Qian-Chao herbal granules on oxalate-induced kidney injury, Bioscience Reports, 39, 2, 2019. Crossref

  51. Grohe Bernd, Taller Adam, Vincent Peter L., Tieu Long D., Rogers Kem A., Heiss Alexander, So̷rensen Esben S., Mittler Silvia, Goldberg Harvey A., Hunter Graeme K., Crystallization of Calcium Oxalates Is Controlled by Molecular Hydrophilicity and Specific Polyanion-Crystal Interactions, Langmuir, 25, 19, 2009. Crossref

  52. Chen Xiong, Zhang Xiao-Bo, Li Dong-Jie, Qi Guan-Nan, Dai Yuan-Qing, Gu Jie, Chen Ming-Quan, Hu Sheng, Liu Zhen-Yu, Yang Zhi-Ming, miR-155 facilitates calcium oxalate crystal-induced HK-2 cell injury via targeting PI3K associated autophagy, Experimental and Molecular Pathology, 115, 2020. Crossref

  53. Pfau Anja, Ermer Theresa, Coca Steven G., Tio Maria Clarissa, Genser Bernd, Reichel Martin, Finkelstein Fredric O., März Winfried, Wanner Christoph, Waikar Sushrut S., Eckardt Kai-Uwe, Aronson Peter S., Drechsler Christiane, Knauf Felix, High Oxalate Concentrations Correlate with Increased Risk for Sudden Cardiac Death in Dialysis Patients, Journal of the American Society of Nephrology, 32, 9, 2021. Crossref

  54. Zhai Wei, Zheng Junhua, Yao Xudong, Peng Bo, Liu Min, Huang Jianhua, Wang Guangchun, Xu Yunfei, Catechin prevents the calcium oxalate monohydrate induced renal calcium crystallization in NRK-52E cells and the ethylene glycol induced renal stone formation in rat, BMC Complementary and Alternative Medicine, 13, 1, 2013. Crossref

  55. Sharbaugh Denise M., Talham Daniel R., Effect of Phospholipase A2 Hydrolysis Products on Calcium Oxalate Precipitation at Lipid Interfaces, Langmuir, 26, 7, 2010. Crossref

  56. Vecitis Chad D., Lesko Timothy, Colussi Agustin J., Hoffmann Michael R., Sonolytic Decomposition of Aqueous Bioxalate in the Presence of Ozone, The Journal of Physical Chemistry A, 114, 14, 2010. Crossref

  57. Cao Lu-Cheng, Honeyman Thomas W., Cooney Rachel, Kennington Lori, Scheid Cheryl R., Jonassen Julie A., Mitochondrial dysfunction is a primary event in renal cell oxalate toxicity, Kidney International, 66, 5, 2004. Crossref

  58. Knight John, Holmes Ross P., Assimos Dean G., Intestinal and renal handling of oxalate loads in normal individuals and stone formers, Urological Research, 35, 3, 2007. Crossref

  59. Kovacevic Larisa, Lu Hong, Kovacevic Natalija, Thomas Ronald, Lakshmanan Yegappan, Cystatin C, Neutrophil Gelatinase-associated Lipocalin, and Lysozyme C: Urinary Biomarkers for Detection of Early Kidney Dysfunction in Children With Urolithiasis, Urology, 143, 2020. Crossref

  60. Gianmoena Kathrin, Gasparoni Nina, Jashari Adelina, Gabrys Philipp, Grgas Katharina, Ghallab Ahmed, Nordström Karl, Gasparoni Gilles, Reinders Jörg, Edlund Karolina, Godoy Patricio, Schriewer Alexander, Hayen Heiko, Hudert Christian A., Damm Georg, Seehofer Daniel, Weiss Thomas S., Boor Peter, Anders Hans-Joachim, Motrapu Manga, Jansen Peter, Schiergens Tobias S., Falk-Paulsen Maren, Rosenstiel Philip, Lisowski Clivia, Salido Eduardo, Marchan Rosemarie, Walter Jörn, Hengstler Jan G., Cadenas Cristina, Epigenomic and transcriptional profiling identifies impaired glyoxylate detoxification in NAFLD as a risk factor for hyperoxaluria, Cell Reports, 36, 8, 2021. Crossref

Статьи, принятые к публикации

PRMT6 promotes the immune evasion of gastric cancer via upregulating ANXA1 Liang Xu, Fenger Zhang, Binqi Yu, Shengnan Jia, Sunfu Fan PURPL promotes M2 macrophage polarization in lung cancer via regulating RBM4/xCT signaling Jipeng Guo, Chongwen Gong, Hao Wang SIAH1 promotes the pyroptosis of cardiomyocytes in diabetic cardiomyopathy via regulating IκB-α/NF-κB signaling Jinbin Wu, Yaoming Yan SLC7A2-mediated lysine catabolism inhibits immunosuppression in triple negative breast cancer Yuanyuan Sun, Yaqing Li, Chengying Jiang, Chenying Liu, Yuanming Song SIAH2-mediated degradation of ACSL4 inhibits the anti-tumor activity of CD8+ T cells in hepatocellular carcinoma Fangzheng Shu, Yuhua Shi, Xiangxiang Shan, Wenzhang Zha, Rengen Fan, Wanjiang Xue RBM15-mediated N6-methyl adenosine (m6A) modification of EZH2 drives the epithelial-mesenchymal transition of cervical cancer Ruixue Wang, Wenhua Tan Evidence-Based Storytelling and A Strategic Roadmap to Promote Cancer Prevention Via Adolescent HPV Vaccination in Northern New England Matthew Dugan, Gary Stein, Jan Carney, Sheila Clifford-Bova KDM4A-AS1 promotes cell proliferation, migration and invasion via the miR-4306/STX6 axis in hepatocellular carcinoma Wei Cao, Yuhan Ren, Ying Liu, Guoshu Cao, Zhen Chen, Fan Wang HDAC1-mediated downregulation of NEU1 exacerbates the aggressiveness of cervical cancer Nanzi Xie, Sisi Mei, Changlan Dai, Wei Chen Effect of miR-26b-5p on progression of acute myeloid leukemia by regulating USP48-mediated Wnt/β-catenin pathway Yu Xie, Lin Tan, Kun Wu, Deyun Li, Chengping Li
Портал Begell Электронная Бибилиотека e-Книги Журналы Справочники и Сборники статей Коллекции Цены и условия подписки Begell House Контакты Language English 中文 Русский Português German French Spain