1
|
Rosas-Martínez L, Rodríguez-Muñoz R, Namorado-Tonix MDC, Missirlis F, Del Valle-Mondragón L, Sánchez-Mendoza A, Reyes-Sánchez JL, Cervantes-Pérez LG. Peroxisome Proliferator-Activated Receptor Alpha Stimulation Preserves Renal Tight Junction Components in a Rat Model of Early-Stage Diabetic Nephropathy. Int J Mol Sci 2024; 25:13152. [PMID: 39684861 DOI: 10.3390/ijms252313152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic hyperglycemia results in morphological and functional alterations of the kidney and microvascular damage, leading to diabetic nephropathy (DN). Since DN progresses to irreversible renal damage, it is important to elucidate a pharmacological strategy aimed for treating DN in the early stage. Here, we used the type 2 diabetic rat model to induce DN and show a nephroprotective effect following the stimulation of PPAR-α, which stabilized renal tight junction components claudin-2, claudin-5, and claudin-16. At 14 weeks old, streptozotocin-induced DN, evidenced by elevated creatinine clearance, proteinuria, and electrolyte excretion, was followed by an elevation in oxidative stress and increasing MMP activities affecting the integrity of claudin-2 and claudin-5. Treatment with a PPAR-α agonists decreased glucose levels in diabetic rats. In addition, we found that the expressions of CLDN-5 in glomeruli, CLDN-2 in proximal tubules, and CLDN-16 in the thick ascending limb of the loop of Henle were increased after treatment. As a result, renal function improved, while the oxidative stress and enzymatic activity of MMP-2 and MMP-9 decreased. In conclusion, PPAR-α stimulation prevented the decrease in claudins through a mechanism involving a correction of hyperglycemia, decreasing it in kidney oxidative stress and MMP-2 and MMP-9 activities, showing a promising nephroprotective action in the early stage of DN.
Collapse
Affiliation(s)
- Lorena Rosas-Martínez
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, Mexico City 14080, Mexico
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico
| | - Rafael Rodríguez-Muñoz
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico
| | - María Del Carmen Namorado-Tonix
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico
| | - Fanis Missirlis
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico
| | - Leonardo Del Valle-Mondragón
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, Mexico City 14080, Mexico
| | - Alicia Sánchez-Mendoza
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, Mexico City 14080, Mexico
| | - José L Reyes-Sánchez
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico
| | - Luz Graciela Cervantes-Pérez
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
2
|
Ramírez Medina CR, Ali I, Baricevic-Jones I, Saleem MA, Whetton AD, Kalra PA, Geifman N. Evaluation of a proteomic signature coupled with the kidney failure risk equation in predicting end stage kidney disease in a chronic kidney disease cohort. Clin Proteomics 2024; 21:34. [PMID: 38762513 PMCID: PMC11102163 DOI: 10.1186/s12014-024-09486-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/25/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND The early identification of patients at high-risk for end-stage renal disease (ESRD) is essential for providing optimal care and implementing targeted prevention strategies. While the Kidney Failure Risk Equation (KFRE) offers a more accurate prediction of ESRD risk compared to static eGFR-based thresholds, it does not provide insights into the patient-specific biological mechanisms that drive ESRD. This study focused on evaluating the effectiveness of KFRE in a UK-based advanced chronic kidney disease (CKD) cohort and investigating whether the integration of a proteomic signature could enhance 5-year ESRD prediction. METHODS Using the Salford Kidney Study biobank, a UK-based prospective cohort of over 3000 non-dialysis CKD patients, 433 patients met our inclusion criteria: a minimum of four eGFR measurements over a two-year period and a linear eGFR trajectory. Plasma samples were obtained and analysed for novel proteomic signals using SWATH-Mass-Spectrometry. The 4-variable UK-calibrated KFRE was calculated for each patient based on their baseline clinical characteristics. Boruta machine learning algorithm was used for the selection of proteins most contributing to differentiation between patient groups. Logistic regression was employed for estimation of ESRD prediction by (1) proteomic features; (2) KFRE; and (3) proteomic features alongside KFRE. RESULTS SWATH maps with 943 quantified proteins were generated and investigated in tandem with available clinical data to identify potential progression biomarkers. We identified a set of proteins (SPTA1, MYL6 and C6) that, when used alongside the 4-variable UK-KFRE, improved the prediction of 5-year risk of ESRD (AUC = 0.75 vs AUC = 0.70). Functional enrichment analysis revealed Rho GTPases and regulation of the actin cytoskeleton pathways to be statistically significant, inferring their role in kidney function and the pathogenesis of renal disease. CONCLUSIONS Proteins SPTA1, MYL6 and C6, when used alongside the 4-variable UK-KFRE achieve an improved performance when predicting a 5-year risk of ESRD. Specific pathways implicated in the pathogenesis of podocyte dysfunction were also identified, which could serve as potential therapeutic targets. The findings of our study carry implications for comprehending the involvement of the Rho family GTPases in the pathophysiology of kidney disease, advancing our understanding of the proteomic factors influencing susceptibility to renal damage.
Collapse
Affiliation(s)
- Carlos Raúl Ramírez Medina
- Stoller Biomarker Discovery Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Ibrahim Ali
- Salford Royal Hospital, Northern Care Alliance Foundation NHS Trust, Salford, UK
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | - Ivona Baricevic-Jones
- Stoller Biomarker Discovery Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Salford Royal Hospital, Northern Care Alliance Foundation NHS Trust, Salford, UK
| | - Moin A Saleem
- Bristol Renal and Children's Renal Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Anthony D Whetton
- Veterinary Health Innovation Engine (vHive), Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Philip A Kalra
- Salford Royal Hospital, Northern Care Alliance Foundation NHS Trust, Salford, UK
| | - Nophar Geifman
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
3
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Cao J, Zhang D, Li W, Yuan W, Luo G, Xie S. Azilsartan improves urinary albumin excretion in hypertension mice. Aging (Albany NY) 2024; 16:4138-4148. [PMID: 38462692 PMCID: PMC10968693 DOI: 10.18632/aging.205271] [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: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 03/12/2024]
Abstract
Hypertension is one of the most important risk factors for chronic kidney diseases, leading to hypertensive nephrosclerosis, including excessive albuminuria. Azilsartan, an angiotensin II type 1 receptor blocker, has been widely used for the treatment of hypertension. However, the effects of Azilsartan on urinary albumin excretion in hypertension haven't been reported before. In this study, we investigated whether Azilsartan possesses a beneficial property against albuminuria in mice treated with angiotensin II and a high-salt diet (ANG/HS). Compared to the control group, the ANG/HS group had higher blood pressure, oxidative stress, and inflammatory response, all of which were rescued by Azilsartan dose-dependently. Importantly, the ANG/HS-induced increase in urinary albumin excretion and decrease in the expression of occludin were reversed by Azilsartan. Additionally, it was shown that increased fluorescence intensity of FITC-dextran, declined trans-endothelial electrical resistance (TEER) values, and reduction of occludin and krüppel-like factor 2 (KLF2) were observed in ANG/HS-treated human renal glomerular endothelial cells (HrGECs), then prevented by Azilsartan. Moreover, the regulatory effect of Azilsartan on endothelial monolayer permeability in ANG/HS-treated HrGECs was abolished by the knockdown of KLF2, indicating KLF2 is required for the effect of Azilsartan. We concluded that Azilsartan alleviated diabetic nephropathy-induced increase in Uterine artery embolization (UAE) mediated by the KLF2/occludin axis.
Collapse
Affiliation(s)
- Jun Cao
- Department of Nephrology, People’s Hospital of Ganzhou, Ganzhou 341001, Jiangxi Province, China
| | - Dandan Zhang
- Department of Nephrology, People’s Hospital of Ganzhou, Ganzhou 341001, Jiangxi Province, China
| | - Wenfeng Li
- Department of Nephrology, People’s Hospital of Ganzhou, Ganzhou 341001, Jiangxi Province, China
| | - Wenjin Yuan
- Department of Nephrology, People’s Hospital of Ganzhou, Ganzhou 341001, Jiangxi Province, China
| | - Gang Luo
- Department of Nephrology, People’s Hospital of Ganzhou, Ganzhou 341001, Jiangxi Province, China
| | - Shaofeng Xie
- Department of Nephrology, People’s Hospital of Ganzhou, Ganzhou 341001, Jiangxi Province, China
| |
Collapse
|
5
|
Duan ZY, Bu R, Liang S, Chen XZ, Zhang C, Zhang QY, Li JJ, Chen XM, Cai GY. Urinary miR-185-5p is a biomarker of renal tubulointerstitial fibrosis in IgA nephropathy. Front Immunol 2024; 15:1326026. [PMID: 38426107 PMCID: PMC10902439 DOI: 10.3389/fimmu.2024.1326026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Background For IgA nephropathy (IgAN), tubular atrophy/interstitial fibrosis is the most important prognostic pathological indicator in the mesangial and endocapillary hypercellularity, segmental sclerosis, interstitial fibrosis/tubular atrophy, and presence of crescents (MEST-C) score. The identification of non-invasive biomarkers for tubular atrophy/interstitial fibrosis would aid clinical monitoring of IgAN progression and improve patient prognosis. Methods The study included 188 patients with primary IgAN in separate confirmation and validation cohorts. The associations of miR-92a-3p, miR-425-5p, and miR-185-5p with renal histopathological lesions and prognosis were explored using Spearman correlation analysis and Kaplan-Meier survival curves. Bioinformatics analysis and dual luciferase experiments were used to identify hub genes for miR-185-5p. The fibrotic phenotypes of tubular epithelial cells were evaluated in vivo and in HK-2 cells. Results miRNA sequencing and cohort validation revealed that the expression levels of miR-92a-3p, miR-425-5p, and miR-185-5p in urine were significantly increased among patients with IgAN; these levels could predict the extent of tubular atrophy/interstitial fibrosis in such patients. The combination of the three biomarkers resulted in an area under the receiver operating characteristic curve of 0.742. The renal prognosis was significantly worse in the miR-185-5p high expression group than in the low expression group (P=0.003). Renal tissue in situ hybridization, bioinformatics analysis, and dual luciferase experiments confirmed that miR-185-5p affects prognosis in patients with IgAN mainly by influencing expression of the target gene tight junction protein 1 (TJP1) in renal tubular epithelial cells. In vitro experiment revealed that an miR-185-5p mimic could reduce TJP1 expression in HK-2 cells, while increasing the levels of α-smooth muscle actin, fibronectin, collagen I, and collagen III; these changes promoted the transformation of renal tubular epithelial cells to a fibrotic phenotype. An miR-185-5p inhibitor can reverse the fibrotic phenotype in renal tubular epithelial cells. In a unilateral ureteral obstruction model, the inhibition of miR-185-5p expression alleviated tubular atrophy/interstitial fibrosis. Conclusion Urinary miR-185-5p, a non-invasive biomarker of tubular atrophy/interstitial fibrosis in IgAN, may promote the transformation of renal tubular epithelial cells to a fibrotic phenotype via TJP1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guang-Yan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| |
Collapse
|
6
|
Mak S, Hammes A. Canonical and Non-Canonical Localization of Tight Junction Proteins during Early Murine Cranial Development. Int J Mol Sci 2024; 25:1426. [PMID: 38338705 PMCID: PMC10855338 DOI: 10.3390/ijms25031426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
This study investigates the intricate composition and spatial distribution of tight junction complex proteins during early mouse neurulation. The analyses focused on the cranial neural tube, which gives rise to all head structures. Neurulation brings about significant changes in the neuronal and non-neuronal ectoderm at a cellular and tissue level. During this process, precise coordination of both epithelial integrity and epithelial dynamics is essential for accurate tissue morphogenesis. Tight junctions are pivotal for epithelial integrity, yet their complex composition in this context remains poorly understood. Our examination of various tight junction proteins in the forebrain region of mouse embryos revealed distinct patterns in the neuronal and non-neuronal ectoderm, as well as mesoderm-derived mesenchymal cells. While claudin-4 exhibited exclusive expression in the non-neuronal ectoderm, we demonstrated a neuronal ectoderm specific localization for claudin-12 in the developing cranial neural tube. Claudin-5 was uniquely present in mesenchymal cells. Regarding the subcellular localization, canonical tight junction localization in the apical junctions was predominant for most tight junction complex proteins. ZO-1 (zona occludens protein-1), claudin-1, claudin-4, claudin-12, and occludin were detected at the apical junction. However, claudin-1 and occludin also appeared in basolateral domains. Intriguingly, claudin-3 displayed a non-canonical localization, overlapping with a nuclear lamina marker. These findings highlight the diverse tissue and subcellular distribution of tight junction proteins and emphasize the need for their precise regulation during the dynamic processes of forebrain development. The study can thereby contribute to a better understanding of the role of tight junction complex proteins in forebrain development.
Collapse
Affiliation(s)
- Shermin Mak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
- Institute for Biology, Free University of Berlin, 14159 Berlin, Germany
| | - Annette Hammes
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
| |
Collapse
|
7
|
Guo C, Jiao M, Cui Y, Li P, Yao J, Dong J, Liao L. Claudin-2 Mediates the Proximal Tubular Epithelial Cell-Fibroblast Crosstalk via Paracrine CTGF. Diabetes Metab Syndr Obes 2024; 17:55-73. [PMID: 38192494 PMCID: PMC10771729 DOI: 10.2147/dmso.s432173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/14/2023] [Indexed: 01/10/2024] Open
Abstract
Purpose Proximal tubular epithelial cell (PTEC) is vulnerable to injury in diabetic kidney disease (DKD) due to high energy expenditure. The injured PTECs-derived profibrotic factors are thought to be driving forces in tubulointerstitial fibrosis (TIF) as they activate surrounding fibroblasts. However, the mechanisms remain unclear. Methods The diabetes with uninephrectomy (DKD) rats were used to evaluated renal histological changes and the expression of Claudin-2 by immunofluorescence staining. Then, Claudin-2 expression in PTECs were modulated and subsequently determined the proliferation and activation of fibroblasts by building a transwell co-culture system in normal glucose (NG)or high glucose (HG) condition. Results Decreased expression of Claudin-2 in PTECs accompanied by tight junction disruption and increased interstitial fibrosis, were detected in DKD rats. In vitro, downregulated Claudin-2 in PTECs promoted proliferation and activation of fibroblasts, which coincided with elevated expression of profibrotic connective tissue growth factor (CTGF) in PTECs. Silenced CTGF inhibited the profibrotic of PTECs via Claudin-2 inhibition. Fibroblasts co-cultured with PTECs transitioned more to myofibroblasts and generated extracellular matrix (ECM) significantly in response to high glucose (HG) stimulation whereas overexpression of Claudin-2 in PTECs reversed the above results. Upregulating CTGF disrupted the beneficial anti-fibrosis effects obtained by overexpression of Claudin-2 in HG condition. Conclusion Our study suggested that Claudin-2 in PTECs, a key mediator of paracellular cation and water transport, promotes the activation and proliferation of surrounding fibroblasts significantly via CTGF in a paracrine manner.
Collapse
Affiliation(s)
- Congcong Guo
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, Shandong, People’s Republic of China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
| | - Yuying Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, Shandong, People’s Republic of China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Pingjiang Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
| | - Jinming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, Shandong, People’s Republic of China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, Shandong, People’s Republic of China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People’s Republic of China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
8
|
Lopes-Gonçalves G, Costa-Pessoa JM, Pimenta R, Tostes AF, da Silva EM, Ledesma FL, Malheiros DMAC, Zatz R, Thieme K, Câmara NOS, Oliveira-Souza M. Evaluation of glomerular sirtuin-1 and claudin-1 in the pathophysiology of nondiabetic focal segmental glomerulosclerosis. Sci Rep 2023; 13:22685. [PMID: 38114708 PMCID: PMC10730508 DOI: 10.1038/s41598-023-49861-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is the leading cause of nephrotic syndrome, which is characterized by podocyte injury. Given that the pathophysiology of nondiabetic glomerulosclerosis is poorly understood and targeted therapies to prevent glomerular disease are lacking, we decided to investigate the tight junction protein claudin-1 and the histone deacetylase sirtuin-1 (SIRT1), which are known to be involved in podocyte injury. For this purpose, we first examined SIRT1, claudin-1 and podocin expression in kidney biopsies from patients diagnosed with nondiabetic FSGS and found that upregulation of glomerular claudin-1 accompanies a significant reduction in glomerular SIRT1 and podocin levels. From this, we investigated whether a small molecule activator of SIRT1, SRT1720, could delay the onset of FSGS in an animal model of adriamycin (ADR)-induced nephropathy; 14 days of treatment with SRT1720 attenuated glomerulosclerosis progression and albuminuria, prevented transcription factor Wilms tumor 1 (WT1) downregulation and increased glomerular claudin-1 in the ADR + SRT1720 group. Thus, we evaluated the effect of ADR and/or SRT1720 in cultured mouse podocytes. The results showed that ADR [1 µM] triggered an increase in claudin-1 expression after 30 min, and this effect was attenuated by pretreatment of podocytes with SRT1720 [5 µM]. ADR [1 µM] also led to changes in the localization of SIRT1 and claudin-1 in these cells, which could be associated with podocyte injury. Although the use of specific agonists such as SRT1720 presents some benefits in glomerular function, their underlying mechanisms still need to be further explored for therapeutic use. Taken together, our data indicate that SIRT1 and claudin-1 are relevant for the pathophysiology of nondiabetic FSGS.
Collapse
Affiliation(s)
- Guilherme Lopes-Gonçalves
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Prof. Lineu Prestes Avenue, Sao Paulo, 05508-000, Brazil.
| | - Juliana Martins Costa-Pessoa
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Prof. Lineu Prestes Avenue, Sao Paulo, 05508-000, Brazil
| | - Ruan Pimenta
- Laboratory of Medical Investigation (LIM 55), Urology Department, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Flavia Tostes
- Laboratory of Neurobiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Eloisa Martins da Silva
- Department of Nephrology, Paulista School of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Roberto Zatz
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Karina Thieme
- Laboratory of Cellular and Molecular Bases of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Nephrology, Paulista School of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Oliveira-Souza
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Prof. Lineu Prestes Avenue, Sao Paulo, 05508-000, Brazil.
| |
Collapse
|
9
|
Lyu CC, Yuan B, Che HY, Meng Y, Zheng Y, He YT, Ji ZH, Cong S, Ji XY, Jiang H, Zhang JB. WITHDRAWN: RNA binding protein Musashi2 regulates dairy cows' mastitis by activating the TGFβ signaling pathway. Int J Biol Macromol 2023:125331. [PMID: 37315671 DOI: 10.1016/j.ijbiomac.2023.125331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
This article has been withdrawn at the request of the editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.
Collapse
Affiliation(s)
- Chen-Chen Lyu
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Bao Yuan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Hao-Yu Che
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Yu Meng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Yi Zheng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Yun-Tong He
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Zhong-Hao Ji
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Shuai Cong
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Xing-Yu Ji
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China
| | - Hao Jiang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China.
| | - Jia-Bao Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China.
| |
Collapse
|
10
|
Wei W, Li W, Yang L, Weeramantry S, Ma L, Fu P, Zhao Y. Tight junctions and acute kidney injury. J Cell Physiol 2023; 238:727-741. [PMID: 36815285 DOI: 10.1002/jcp.30976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/24/2023]
Abstract
Acute kidney injury (AKI) is characterized by a rapid reduction in kidney function caused by various etiologies. Tubular epithelial cell dysregulation plays a pivotal role in the pathogenesis of AKI. Tight junction (TJ) is the major molecular structure that connects adjacent epithelial cells and is critical in maintaining barrier function and determining the permeability of epithelia. TJ proteins are dysregulated in various types of AKI, and some reno-protective drugs can reverse TJ changes caused by insult. An in-depth understanding of TJ regulation and its causality with AKI will provide more insight to the disease pathogenesis and will shed light on the potential role of TJs to serve as novel therapeutic targets in AKI.
Collapse
Affiliation(s)
- Wei Wei
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiying Li
- Department of Internal Medicine, Florida Hospital/AdventHealth, Orlando, Florida, USA
| | - Letian Yang
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Savidya Weeramantry
- Department of Internal Medicine, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Liang Ma
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Fu
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuliang Zhao
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Hurtado KA, Janda J, Schnellmann RG. Lasmiditan promotes recovery from acute kidney injury through induction of mitochondrial biogenesis. Am J Physiol Renal Physiol 2023; 324:F56-F63. [PMID: 36326468 PMCID: PMC9762961 DOI: 10.1152/ajprenal.00249.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Acute kidney injury (AKI) involves rapid loss of renal function and occurs in 8-16% of hospitalized patients. AKI can be induced by drugs, sepsis, and ischemia-reperfusion (I/R). Hallmarks of AKI include mitochondrial and microvasculature dysfunction as well as renal tubular injury. There is currently no available therapeutic for AKI. Previously, our group identified that serotonin (5-HT)1F receptor agonism with lasmiditan accelerated endothelial cell recovery and induced mitochondrial biogenesis (MB) in vitro. We hypothesized that lasmiditan, a Federal Drug Administration-approved drug, would induce MB and improve microvascular and renal function in a mouse model of AKI. Male mice were subjected to renal I/R and treated with lasmiditan (0.3 mg/kg) or vehicle beginning 24 h after injury and then daily until euthanasia at 6 or 12 days. Serum creatinine was measured to estimate glomerular filtration rate. The renal cortex was assessed for mitochondrial density, vascular permeability and integrity, tubular damage, and interstitial fibrosis. Lasmiditan increased mitochondrial number (1.4-fold) in renal cortices. At 6 days, serum creatinine decreased 41% in the I/R group and 72% with lasmiditan. At 6 or 12 days, kidney injury molecule-1 increased in the I/R group and decreased 50% with lasmiditan. At 12 days, interstitial fibrosis decreased with lasmiditan by 50% and collagen type 1 by 38%. Evan's blue dye leakage increased 2.5-fold in the I/R group and was restored with lasmiditan. The tight junction proteins zonula occludens-1, claudin-2, and claudin-5 decreased in the I/R group and recovered with lasmiditan. At 6 or 12 days, peroxisome proliferator-activated receptor-γ coactivator-1α and electron transport chain complexes increased only with lasmiditan. In conclusion, lasmiditan treatment beginning AKI induces MB, attenuated vascular and tubular injury, decreased interstitial fibrosis, and lowered serum creatinine. Given that lasmiditan is a Federal Drug Administration-approved drug, these preclinical data support repurposing lasmiditan as a therapeutic for AKI.NEW & NOTEWORTHY AKI pathology involves a rapid decline in kidney function and occurs in 8-16% of hospitalized patients. There is currently no therapeutic for AKI. AKI results in mitochondria dysfunction, microvasculature injury, and loss of renal tubular function. In an I/R-induced AKI mouse model, treatment with the FDA-approved 5-HT1F receptor-selective agonist lasmiditan induced mitochondrial biogenesis, improved vascular integrity, reduced fibrosis, and reduced proximal tubule damage. These data support repurposing lasmiditan for the treatment of AKI.
Collapse
Affiliation(s)
- Kevin A Hurtado
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
- Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
- Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
12
|
Vallorz EL, Janda J, Mansour HM, Schnellmann RG. Kidney targeting of formoterol containing polymeric nanoparticles improves recovery from ischemia reperfusion-induced acute kidney injury in mice. Kidney Int 2022; 102:1073-1089. [PMID: 35779607 DOI: 10.1016/j.kint.2022.05.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/22/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022]
Abstract
The β2 adrenergic receptor agonist, formoterol, is an inducer of mitochondrial biogenesis and restorer of mitochondrial and kidney function in acute and chronic models of kidney injury. Unfortunately, systemic administration of formoterol has the potential for adverse cardiovascular effects, increased heart rate, and decreased blood pressure. To minimize these effects, we developed biodegradable and biocompatible polymeric nanoparticles containing formoterol that target the kidney, thereby decreasing the effective dose, and lessen cardiovascular effects while restoring kidney function after injury. Male C57Bl/6 mice, treated with these nanoparticles daily, had reduced ischemia-reperfusion-induced serum creatinine and kidney cortex kidney injury molecule-1 levels by 78% and 73% respectively, compared to control mice six days after injury. With nanoparticle therapy, kidney cortical mitochondrial number and proteins reduced by ischemic injury, recovered to levels of sham-operated mice. Tubular necrosis was reduced 69% with nanoparticles treatment. Nanoparticles improved kidney recovery even when the dosing frequency was reduced from daily to two days per week. Finally, compared to treatment with formoterol-free drug alone, these nanoparticles did not increase heart rate nor decrease blood pressure. Thus, targeted kidney delivery of formoterol-containing nanoparticles is an improvement in standard formoterol therapy for ischemia-reperfusion-induced acute kidney injuries by decreasing the dose, dosing frequency, and cardiac side effects.
Collapse
Affiliation(s)
- Ernest L Vallorz
- Department of Pharmacology and Toxicology, The University of Arizona R. Ken Coit College of Pharmacy, Skaggs Pharmaceutical Sciences Center, Tucson, Arizona, USA
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, The University of Arizona R. Ken Coit College of Pharmacy, Skaggs Pharmaceutical Sciences Center, Tucson, Arizona, USA
| | - Heidi M Mansour
- Department of Pharmacology and Toxicology, The University of Arizona R. Ken Coit College of Pharmacy, Skaggs Pharmaceutical Sciences Center, Tucson, Arizona, USA; The University of Arizona College of Medicine, Tucson, Arizona, USA; The University of Arizona, BIO5 Institute, Tucson, Arizona, USA
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, The University of Arizona R. Ken Coit College of Pharmacy, Skaggs Pharmaceutical Sciences Center, Tucson, Arizona, USA; The University of Arizona College of Medicine, Tucson, Arizona, USA; The University of Arizona, BIO5 Institute, Tucson, Arizona, USA; Southern Arizona VA Health Care System, USA.
| |
Collapse
|
13
|
Naryzhny S, Legina O. Zonulin — regulation of tight contacts in the brain and intestine — facts and hypotheses. BIOMEDITSINSKAYA KHIMIYA 2022; 68:309-320. [DOI: 10.18097/pbmc20226805309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In recent years, the interrelationship between the brain and the gut has become an area of high scientific interest. The intestine is responsible not only for digestion, as it contains millions of neurons, its own immune system, and affects the emotional and cognitive processes. The relationship between the gut and the brain suggests that the processes carried out by the gut microbiota play a significant role in the regulation of brain function, and vice versa. A special role here is played by intercellular tight junctions (TJ), where the zonulin protein holds an important place. Zonulin, an unprocessed precursor of mature haptoglobin, is the only physiological modulator of intercellular TJ that can reversibly regulate the permeability of the intestinal (IB) and blood-brain (BBB) barriers in the human body. BBB disruption and altered microbiota composition are associated with many diseases, including neurological disorders and neuroinflammation. That is, there is a gut-brain axis (GBA) — a communication system through which the brain modulates the functions of the gastrointestinal tract (GIT) and vice versa. GBA is based on neuronal, endocrine, and immunological mechanisms that are interconnected at the organismal, organ, cellular, and molecular levels.
Collapse
Affiliation(s)
- S.N. Naryzhny
- Institute of Biomedical Chemistry, Moscow, Russia; Petersburg Institute of Nuclear Physics B.P. Konstantinova National Research Center “Kurchatov Institute”, Gatchina, Russia
| | - O.K. Legina
- Petersburg Institute of Nuclear Physics B.P. Konstantinova National Research Center “Kurchatov Institute”, Gatchina, Russia
| |
Collapse
|
14
|
Chmiel TA, Gardel ML. Confluence and tight junction dependence of volume regulation in epithelial tissue. Mol Biol Cell 2022; 33:ar98. [PMID: 35731553 PMCID: PMC9582799 DOI: 10.1091/mbc.e22-03-0073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/11/2022] Open
Abstract
Epithelial cell volume regulation is a key component to tissue stability and dynamics. In particular, how cells respond to osmotic stresses is of significant physiological interest in kidney epithelial tissue. For individual mammalian cells, it is well established that Na-K-2Cl cotransporter (NKCC) channels mediate cell volume homeostasis in response to hyperosmotic stress. However, whether mature epithelium responds similarly is not well known. Here we show that while small colonies of madin darby canine kidney (MDCK) epithelial cells behave similarly to single cells and exhibit volume homeostasis that is dependent on the NKCC channel function, mature epithelial tissue does not. Instead, the cell volume decreases by 33% when confluent monolayers or acini formed from MDCK cells are subjected to hyperosmotic stress. We show that the tight junction protein zonula occludins-1 (ZO-1), and Rho-associated kinase (ROCK) are essential for osmotic regulation of cell volume in mature epithelium. Because these both are known to be essential for tight junction assembly, this strongly suggests a role for tight junctions in changing volume response in mature epithelium. Thus, tight junctions act either directly or indirectly in osmotic pressure response of epithelial tissue to suppress volume homeostasis common to isolated epithelial cells.
Collapse
Affiliation(s)
- Theresa A. Chmiel
- Institute for Biophysical Dynamics, James Franck Institute, Department of Physics, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| | - Margaret L. Gardel
- Institute for Biophysical Dynamics, James Franck Institute, Department of Physics, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| |
Collapse
|
15
|
Hansen J, Sealfon R, Menon R, Eadon MT, Lake BB, Steck B, Anjani K, Parikh S, Sigdel TK, Zhang G, Velickovic D, Barwinska D, Alexandrov T, Dobi D, Rashmi P, Otto EA, Rivera M, Rose MP, Anderton CR, Shapiro JP, Pamreddy A, Winfree S, Xiong Y, He Y, de Boer IH, Hodgin JB, Barisoni L, Naik AS, Sharma K, Sarwal MM, Zhang K, Himmelfarb J, Rovin B, El-Achkar TM, Laszik Z, He JC, Dagher PC, Valerius MT, Jain S, Satlin LM, Troyanskaya OG, Kretzler M, Iyengar R, Azeloglu EU. A reference tissue atlas for the human kidney. SCIENCE ADVANCES 2022; 8:eabn4965. [PMID: 35675394 PMCID: PMC9176741 DOI: 10.1126/sciadv.abn4965] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/20/2022] [Indexed: 05/08/2023]
Abstract
Kidney Precision Medicine Project (KPMP) is building a spatially specified human kidney tissue atlas in health and disease with single-cell resolution. Here, we describe the construction of an integrated reference map of cells, pathways, and genes using unaffected regions of nephrectomy tissues and undiseased human biopsies from 56 adult subjects. We use single-cell/nucleus transcriptomics, subsegmental laser microdissection transcriptomics and proteomics, near-single-cell proteomics, 3D and CODEX imaging, and spatial metabolomics to hierarchically identify genes, pathways, and cells. Integrated data from these different technologies coherently identify cell types/subtypes within different nephron segments and the interstitium. These profiles describe cell-level functional organization of the kidney following its physiological functions and link cell subtypes to genes, proteins, metabolites, and pathways. They further show that messenger RNA levels along the nephron are congruent with the subsegmental physiological activity. This reference atlas provides a framework for the classification of kidney disease when multiple molecular mechanisms underlie convergent clinical phenotypes.
Collapse
Affiliation(s)
- Jens Hansen
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Sealfon
- Princeton University, Princeton, NJ, USA
- Flatiron Institute, New York, NY, USA
| | - Rajasree Menon
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | | | - Blue B. Lake
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Becky Steck
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kavya Anjani
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Samir Parikh
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Tara K. Sigdel
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Guanshi Zhang
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
| | | | - Daria Barwinska
- Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Dejan Dobi
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Priyanka Rashmi
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Edgar A. Otto
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Miguel Rivera
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Michael P. Rose
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Christopher R. Anderton
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - John P. Shapiro
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Annapurna Pamreddy
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
| | - Seth Winfree
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuguang Xiong
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yongqun He
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Ian H. de Boer
- Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
| | | | | | - Abhijit S. Naik
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kumar Sharma
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
| | - Minnie M. Sarwal
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Jonathan Himmelfarb
- Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
| | - Brad Rovin
- Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Zoltan Laszik
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | | | | | - M. Todd Valerius
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Sanjay Jain
- Washington University in Saint Louis School of Medicine, St. Louis, MS, USA
| | - Lisa M. Satlin
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olga G. Troyanskaya
- Princeton University, Princeton, NJ, USA
- Flatiron Institute, New York, NY, USA
| | | | - Ravi Iyengar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Kidney Precision Medicine Project
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Princeton University, Princeton, NJ, USA
- Flatiron Institute, New York, NY, USA
- University of Michigan School of Medicine, Ann Arbor, MI, USA
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- University of California San Francisco School of Medicine, San Francisco, CA, USA
- Ohio State University College of Medicine, Columbus, OH, USA
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
- Pacific Northwest National Laboratory, Richland, WA, USA
- European Molecular Biology Laboratory, Heidelberg, Germany
- Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
- Duke University School of Medicine, Durham, NC, USA
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- Washington University in Saint Louis School of Medicine, St. Louis, MS, USA
| |
Collapse
|
16
|
Jo CH, Kim S, Kim GH. Claudins in kidney health and disease. Kidney Res Clin Pract 2022; 41:275-287. [PMID: 35354245 PMCID: PMC9184838 DOI: 10.23876/j.krcp.21.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/29/2021] [Indexed: 11/04/2022] Open
Abstract
Claudins are strategically located to exert their physiologic actions along with the nephron segments from the glomerulus. Claudin-1 is normally located in the Bowman’s capsule, but its overexpression can reach the podocytes and lead to albuminuria. In the proximal tubule (PT), claudin-2 forms paracellular channels selective for water, Na+, K+, and Ca2+. Claudin-2 gene mutations are associated with hypercalciuria and kidney stones. Claudin-10 has two splice variants, -10a and -10b; Claudin-10a acts as an anion-selective channel in the PT, and claudin-10b functions as a cation-selective pore in the thick ascending limb (TAL). Claudin-16 and claudin-19 mediate paracellular transport of Na+, Ca2+, and Mg2+ in the TAL, where the expression of claudin-3/16/19 and claudin-10b are mutually exclusive. The claudin-16 or -19 mutation causes familial hypomagnesemia with hypercalciuria and nephrocalcinosis. Claudin-14 polymorphisms have been linked to increased risk of hypercalciuria. Claudin-10b mutations produce HELIX syndrome, which encompasses hypohidrosis, electrolyte imbalance, lacrimal gland dysfunction, ichthyosis, and xerostomia. Hypercalciuria and magnesuria in metabolic acidosis are related to downregulation of PT and TAL claudins. In the TAL, stimulation of calcium-sensing receptors upregulates claudin-14 and negatively acts on the claudin-16/19 complex. Claudin-3 acts as a general barrier to ions in the collecting duct. If this barrier is disturbed, urine acidification might be impaired. Claudin-7 forms a nonselective paracellular channel facilitating Cl– and Na+ reabsorption in the collecting ducts. Claudin-4 and -8 serve as anion channels and mediate paracellular Cl– transport; their upregulation may contribute to pseudohypoaldosteronism II and salt-sensitive hypertension.
Collapse
Affiliation(s)
- Chor ho Jo
- Hanyang Biomedical Research Institute, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Sua Kim
- Hanyang Biomedical Research Institute, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Gheun-Ho Kim
- Hanyang Biomedical Research Institute, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
- Correspondence: Gheun-Ho Kim Department of Internal Medicine, Hanyang University College of Medicine, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea. E-mail:
| |
Collapse
|
17
|
Xiao M, Zheng L, Zhang X, Duan X, Hang T, Lu S, Liu S, Lin H. Renal-on-Chip Microfluidic Platform with a Force-Sensitive Resistor (ROC-FS) for Molecular Pathogenesis Analysis of Hydronephrosis. Anal Chem 2021; 94:748-757. [PMID: 34951537 DOI: 10.1021/acs.analchem.1c03155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydronephrosis is one of the most common diseases in urology. However, due to the difficulties in clinical trials and the lack of reliable in vitro platforms, the surgical indicators are not clear. Herein, the renal-on-chip with a force-sensitive resistor microfluidic platform was established to simulate the state of hydronephrosis. Cell counting kit-8 (CCK-8) and tight junction protein claudin-2 were detected on a renal-on-chip microfluidic platform with a force-sensitive resistor (ROC-FS). The results indicated that the ROC-FS had normal physiological functions and the cell viability on ROC-FS declined to around 40% after 48 h of hydronephrosis-simulated treatment. In addition, proteomics analysis of 15 clinical ureteropelvic junction obstruction (UPJO) samples showed that compared with normal children, a total of 50 common proteins were differentially expressed in UPJO children (P < 0.05, |log2fold change| ≥ 1). Metabolomic analysis of 39 clinical UPJO samples showed that a total of 241 metabolisms were dysregulated. Subsequent immunofluorescence and enzyme-linked immunosorbent assay (ELISA) analysis using ROC-FS were performed to identify the clinical multi-omics results for screening. All results pointed out that the TGF-β-related signaling pathways and arginine-related metabolism signaling pathways were dysregulated and α-SMA, AGT, and AGA might be the potential biomarkers of hydronephrosis. In addition, correlation analysis of AGT and KLK1 with differential renal function (DRF) from clinical samples indicated good correlation coefficients (R2 0.923, 0.8742, 0.6412, and 0.8347). This demonstrates the state of hydronephrosis could be significantly correlated with the biomarkers. These findings could provide a reliable reference for determining surgical biomarkers clinically, and ROC could be further used in the analysis of other kidney diseases.
Collapse
Affiliation(s)
- Mingming Xiao
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Lulu Zheng
- Engineering Research Center of Optical Instrument and System, Ministry of Education, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, 526 Jugong Road, Shanghai 200093, China
| | - Xinlian Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Xiaoxiao Duan
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Tian Hang
- Department of Pediatric Surgery, Jiaxing Women and Children Hospital Affiliated to Jiaxing University, 2468 East Zhonghuan Road, Jiaxing, Zhejiang 314050, China
| | - Shijiao Lu
- Department of Pediatric Surgery, Jiaxing Women and Children Hospital Affiliated to Jiaxing University, 2468 East Zhonghuan Road, Jiaxing, Zhejiang 314050, China
| | - Sixiu Liu
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Houwei Lin
- Department of Pediatric Surgery, Jiaxing Women and Children Hospital Affiliated to Jiaxing University, 2468 East Zhonghuan Road, Jiaxing, Zhejiang 314050, China
| |
Collapse
|
18
|
Rzhevskaya ON, Moiseeva AY, Esaulenko AN, Pinchuk AV, Alidzhanova KG. Chronic kidney disease as a risk factor for acute stroke. TRANSPLANTOLOGIYA. THE RUSSIAN JOURNAL OF TRANSPLANTATION 2021; 13:382-397. [DOI: 10.23873/2074-0506-2021-13-4-382-397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
One of the most relevant issues of nephrology, neurology, and cardiology is the management and treatment of patients with chronic kidney disease and stroke. Patients with chronic kidney disease have a risk of both thrombotic complications and bleeding, and they have a high risk of both ischemic and hemorrhagic stroke. Chronic kidney disease significantly worsens the outcome of stroke by limiting the treatment due to reduced drug clearance and side effects. Hemodialysis which causes drastic hemodynamic and biochemical changes leads to the "stress" of the cerebral vascular system, increasing the risk of stroke; kidney transplantation reduces the risk of stroke due to functional recovery. Chronic kidney disease and stroke have significant socio-economic consequences. Patients with end-stage chronic kidney disease, as a rule, are not included in clinical trials; and stroke treatment tactics have not been developed for them. This review examines the interaction between kidneys and brain, the pathophysiology and epidemiology of stroke in all stages of chronic kidney disease, after kidney transplantation and discusses the management and treatment of chronic kidney disease patients with stroke.The investigation of the factors responsible for the high prevalence of brain lesions in chronic kidney disease will allow developing new treatment methods.
Collapse
Affiliation(s)
- O. N. Rzhevskaya
- N.V. Sklifosovsky Research Institute for Emergency Medicine; Department of Transplantology and Artificial Organs, A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - A. Y. Moiseeva
- N.V. Sklifosovsky Research Institute for Emergency Medicine
| | | | - A. V. Pinchuk
- N.V. Sklifosovsky Research Institute for Emergency Medicine; Department of Transplantology and Artificial Organs, A.I. Yevdokimov Moscow State University of Medicine and Dentistry; Research Institute for Healthcare Organization and Medical Management
| | | |
Collapse
|
19
|
Kan X, Liu J, Cai X, Huang Y, Xu P, Fu S, Guo W, Hu G. Tartary buckwheat flavonoids relieve the tendency of mammary fibrosis induced by HFD during pregnancy and lactation. Aging (Albany NY) 2021; 13:25377-25392. [PMID: 34890369 PMCID: PMC8714130 DOI: 10.18632/aging.203752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
Mammary gland fibrosis is a chronic and irreversible disease. Tartary buckwheat flavonoids (TBF) are a natural product of flavonoid extracts from buckwheat and have a wide range of biological activities. The purpose of this experiment was to explore whether HFD during pregnancy and lactation induces fibrosis of the mammary tissue and whether TBF alleviates the damage caused by HFD, along with its underlying mechanism. The HFD significantly increased the levels of TNF-α, IL-6, IL-1β, and MPO; significantly damaged the integrity of the blood-milk barrier; significantly increased the levels of collagen 1, vimentin and α-SMA, and reduced the level of E-cadherin. However, these effects were alleviated by TBF. Mechanistic studies showed that TBF inhibited the activation of AKT/NF-κB signaling and predicted the AKT amino acid residues that formed hydrogen bonds with TBF; in addition, these studies not only revealed that TBF promoted the expression of the tight junction proteins (TJs) claudin-3, occludin and ZO-1 and inhibited the activation of TGF-β/Smad signaling but also predicted the Smad MH2 amino acid residues that formed hydrogen bonds with TBF. Conclusion: HFD consumption during pregnancy and lactation induced the tendency of mammary fibrosis. TBF alleviated the tendency of mammary fibrosis by inhibiting the activation of AKT/NF-κB, repairing the blood-milk barrier and inhibiting the activation of TGF-β/Smad signaling.
Collapse
Affiliation(s)
- Xingchi Kan
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Juxiong Liu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiangyu Cai
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yaping Huang
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Ping Xu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shoupeng Fu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Wenjin Guo
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guiqiu Hu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
20
|
Hussain B, Kasinath V, Madsen JC, Bromberg J, Tullius SG, Abdi R. Intra-Organ Delivery of Nanotherapeutics for Organ Transplantation. ACS NANO 2021; 15:17124-17136. [PMID: 34714050 PMCID: PMC9050969 DOI: 10.1021/acsnano.1c04707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Targeted delivery of therapeutics through the use of nanoparticles (NPs) has emerged as a promising method that increases their efficacy and reduces their side effects. NPs can be tailored to localize to selective tissues through conjugation to ligands that bind cell-specific receptors. Although the vast majority of nanodelivery platforms have focused on cancer therapy, efforts have begun to introduce nanotherapeutics to the fields of immunology as well as transplantation. In this review, we provide an overview from a clinician's perspective of current nanotherapeutic strategies to treat solid organ transplants with NPs during the time interval between organ harvest from the donor and placement into the recipient, an innovative technology that can provide major benefits to transplant patients. The use of ex vivo normothermic machine perfusion (NMP), which is associated with preserving the function of the organ following transplantation, also provides an ideal opportunity for a localized, sustained, and controlled delivery of nanotherapeutics to the organ during this critical time period. Here, we summarize previous endeavors to improve transplantation outcomes by treating the organ with NPs prior to placement in the recipient. Investigations in this burgeoning field of research are promising, but more extensive studies are needed to overcome the physiological challenges to achieving effective nanotherapeutic delivery to transplanted organs discussed in this review.
Collapse
Affiliation(s)
- Bilal Hussain
- Transplantation Research Center and Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Vivek Kasinath
- Transplantation Research Center and Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Joren C. Madsen
- Department of Surgery and Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jonathan Bromberg
- Departments of Surgery and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Stefan G. Tullius
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Reza Abdi
- Transplantation Research Center and Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
21
|
Wang B, Qian JY, Tang TT, Lin LL, Yu N, Guo HL, Ni WJ, Lv LL, Wen Y, Li ZL, Wu M, Cao JY, Liu BC. VDR/Atg3 Axis Regulates Slit Diaphragm to Tight Junction Transition via p62-Mediated Autophagy Pathway in Diabetic Nephropathy. Diabetes 2021; 70:2639-2651. [PMID: 34376476 DOI: 10.2337/db21-0205] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/01/2021] [Indexed: 12/09/2022]
Abstract
Foot process effacement is an important feature of early diabetic nephropathy (DN), which is closely related to the development of albuminuria. Under certain nephrotic conditions, the integrity and function of the glomerular slit diaphragm (SD) structure were impaired and replaced by the tight junction (TJ) structure, resulting in so-called SD-TJ transition, which could partially explain the effacement of foot processes at the molecular level. However, the mechanism underlying the SD-TJ transition has not been described in DN. Here, we demonstrated that impaired autophagic flux blocked p62-mediated degradation of ZO-1 (TJ protein) and promoted podocytes injury via activation of caspase3 and caspase8. Interestingly, the expression of VDR in podocytes was decreased under diabetes conditions, which impaired autophagic flux through downregulating Atg3. Of note, we also found that VDR abundance was negatively associated with impaired autophagic flux and SD-TJ transition in the glomeruli from human renal biopsy samples with DN. Furthermore, VDR activation improved autophagic flux and attenuated SD-TJ transition in the glomeruli of diabetic animal models. In conclusion, our data provided the novel insight that VDR/Atg3 axis deficiency resulted in SD-TJ transition and foot processes effacement via blocking the p62-mediated autophagy pathway in DN.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Jing-Yi Qian
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Li-Lu Lin
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Nan Yu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Hong-Lei Guo
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei-Jie Ni
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Ling-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Min Wu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
22
|
Llorens S, Nava E, Muñoz-López M, Sánchez-Larsen Á, Segura T. Neurological Symptoms of COVID-19: The Zonulin Hypothesis. Front Immunol 2021; 12:665300. [PMID: 33981312 PMCID: PMC8107207 DOI: 10.3389/fimmu.2021.665300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
The irruption of SARS-CoV-2 during 2020 has been of pandemic proportions due to its rapid spread and virulence. COVID-19 patients experience respiratory, digestive and neurological symptoms. Distinctive symptom as anosmia, suggests a potential neurotropism of this virus. Amongst the several pathways of entry to the nervous system, we propose an alternative pathway from the infection of the gut, involving Toll-like receptor 4 (TLR4), zonulin, protease-activated receptor 2 (PAR2) and zonulin brain receptor. Possible use of zonulin antagonists could be investigated to attenuate neurological manifestations caused by SARS-CoV-19 infection.
Collapse
Affiliation(s)
- Sílvia Llorens
- Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha, Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), University of Castilla-La Mancha, Albacete, Spain
| | - Eduardo Nava
- Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha, Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), University of Castilla-La Mancha, Albacete, Spain
| | - Mónica Muñoz-López
- Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha, Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), University of Castilla-La Mancha, Albacete, Spain
| | | | - Tomás Segura
- Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha, Albacete, Spain.,Servicio de Neurología, Hospital General Universitario de Albacete, Albacete, Spain.,Instituto de Investigación en Discapacidades Neurológicas (IDINE), University of Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
23
|
Mendoza C, Nagidi SH, Mizrachi D. Molecular Characterization of the Extracellular Domain of Human Junctional Adhesion Proteins. Int J Mol Sci 2021; 22:ijms22073482. [PMID: 33801758 PMCID: PMC8037251 DOI: 10.3390/ijms22073482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/22/2022] Open
Abstract
The junction adhesion molecule (JAM) family of proteins play central roles in the tight junction (TJ) structure and function. In contrast to claudins (CLDN) and occludin (OCLN), the other membrane proteins of the TJ, whose structure is that of a 4α-helix bundle, JAMs are members of the immunoglobulin superfamily. The JAM family is composed of four members: A, B, C and 4. The crystal structure of the extracellular domain of JAM-A continues to be used as a template to model the secondary and tertiary structure of the other members of the family. In this article, we have expressed the extracellular domains of JAMs fused with maltose-binding protein (MBP). This strategy enabled the work presented here, since JAM-B, JAM-C and JAM4 are more difficult targets due to their more hydrophobic nature. Our results indicate that each member of the JAM family has a unique tertiary structure in spite of having similar secondary structures. Surface plasmon resonance (SPR) revealed that heterotypic interactions among JAM family members can be greatly favored compared to homotypic interactions. We employ the well characterized epithelial cadherin (E-CAD) as a means to evaluate the adhesive properties of JAMs. We present strong evidence that suggests that homotypic or heterotypic interactions among JAMs are stronger than that of E-CADs.
Collapse
Affiliation(s)
- Christopher Mendoza
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
| | - Sai Harsha Nagidi
- Department of Molecular Microbiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
| | - Dario Mizrachi
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
- Correspondence:
| |
Collapse
|
24
|
Rosas-Martínez L, Rodríguez-Muñoz R, Namorado-Tonix MDC, Missirlis F, Del Valle-Mondragón L, Sánchez-Mendoza A, Reyes-Sánchez JL, Cervantes-Pérez LG. Hyperglycemic levels in early stage of diabetic nephropathy affect differentially renal expression of claudins-2 and -5 by oxidative stress. Life Sci 2021; 268:119003. [PMID: 33417957 DOI: 10.1016/j.lfs.2020.119003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/17/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023]
Abstract
AIMS This study attempts to elicit whether the level of hyperglycemia in an early stage of diabetic nephropathy changes the renal expression of claudins-2 and -5 and to determine the involvement of glucose-induced oxidative stress. MAIN METHODS Streptozotocin-induced type-1 and type-2 diabetic (DM1, DM2)-rat models were used. At 14-week old, the rats were placed in metabolic cages to evaluate proteinuria, creatinine clearance, and electrolyte excretion. Proximal tubules and glomeruli were isolated and analyzed by Western blot and immunofluorescence. Renal oxidative stress and metalloproteinase activities were evaluated. KEY FINDINGS We found that claudin-5 expression in glomeruli and claudin-2 expression in proximal tubules were significantly reduced in DM1 versus DM2 model, paralleling with higher proteinuria and loss of sodium and potassium reabsorption, increased malondialdehyde levels, but lower antioxidant capacity in both models. Enzymatic activity of MMP-2 and-9 was increased in both diabetic groups versus control being higher in DM1 than DM2, suggesting higher claudin's degradation. SIGNIFICANCE The level of hyperglycemia determines the time-dependent progression to diabetic nephropathy; hyperglycemia-induced oxidative stress parallels an increase in metalloproteinases (MMPs) activities consequently affecting the integrity of claudin-2 and -5 in glomerulus and proximal tubule. Our results suggest that chronic high-glycemia levels in early stages of diabetic nephropathy decrease expression of claudins-2 and -5, increase oxidative stress, and induce MMP-activity faster than chronic middle-glycemia levels.
Collapse
Affiliation(s)
- Lorena Rosas-Martínez
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, 14080 Mexico City, Mexico; Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politecnico Nacional, 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Mexico City, Mexico.
| | - Rafael Rodríguez-Muñoz
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politecnico Nacional, 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Mexico City, Mexico.
| | - María Del Carmen Namorado-Tonix
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politecnico Nacional, 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Mexico City, Mexico
| | - Fanis Missirlis
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politecnico Nacional, 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Mexico City, Mexico.
| | - Leonardo Del Valle-Mondragón
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, 14080 Mexico City, Mexico
| | - Alicia Sánchez-Mendoza
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, 14080 Mexico City, Mexico
| | - José L Reyes-Sánchez
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of National Polytechnic Institute, CINVESTAV-IPN, Instituto Politecnico Nacional, 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Mexico City, Mexico.
| | - Luz Graciela Cervantes-Pérez
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No. 1, Col. Seccion XVI, Tlalpan, 14080 Mexico City, Mexico.
| |
Collapse
|
25
|
Liu H, Yan R, Liang L, Zhang H, Xiang J, Liu L, Zhang X, Mao Y, Peng W, Xiao Y, Zhang F, Zhou Y, Shi M, Wang Y, Guo B. The role of CDX2 in renal tubular lesions during diabetic kidney disease. Aging (Albany NY) 2021; 13:6782-6803. [PMID: 33621200 PMCID: PMC7993706 DOI: 10.18632/aging.202537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/09/2020] [Indexed: 02/05/2023]
Abstract
Renal tubules are vulnerable targets of various factors causing kidney injury in diabetic kidney disease (DKD), and the degree of tubular lesions is closely related to renal function. Abnormal renal tubular epithelial cells (RTECs) differentiation and depletion of cell junction proteins are important in DKD pathogenesis. Caudal-type homeobox transcription factor 2 (CDX2), represents a key nuclear transcription factor that maintains normal proliferation and differentiation of the intestinal epithelium. The present study aimed to evaluate the effects of CDX2 on RTECs differentiation and cell junction proteins in DKD. The results demonstrated that CDX2 was mainly localized in renal tubules, and downregulated in various DKD models. CDX2 upregulated E-cadherin and suppressed partial epithelial-mesenchymal transition (EMT), which can alleviate hyperglycemia-associated RTECs injury. Cystic fibrosis transmembrane conductance regulator (CFTR) was regulated by CDX2 in NRK-52E cells, and CFTR interfered with β-catenin activation by binding to Dvl2, which is an essential component of Wnt/β-catenin signaling. CFTR knockdown abolished the suppressive effects of CDX2 on Wnt/β-catenin signaling, thereby upregulating cell junction proteins and inhibiting partial EMT in RTECs. In summary, CDX2 can improve renal tubular lesions during DKD by increasing CFTR amounts to suppress the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Huiming Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rui Yan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Luqun Liang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Huifang Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Jiayi Xiang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Xiaohuan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yanwen Mao
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Wei Peng
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yuxia Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| |
Collapse
|
26
|
Kang DH. Loosening of the mesothelial barrier as an early therapeutic target to preserve peritoneal function in peritoneal dialysis. Kidney Res Clin Pract 2020; 39:136-144. [PMID: 32576713 PMCID: PMC7321674 DOI: 10.23876/j.krcp.20.052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/21/2022] Open
Abstract
Phenotype transition of peritoneal mesothelial cells (MCs) including the epithelial-to-mesenchymal transition (EMT) is regarded as an early mechanism of peritoneal dysfunction and fibrosis in peritoneal dialysis (PD), producing proinflammatory and pro-fibrotic milieu in the intra-peritoneal cavity. Loosening of intercellular tight adhesion between adjacent MCs as an initial process of EMT creates the environment where mesothelium and submesothelial tissue are more vulnerable to the composition of bio-incompatible dialysates, reactive oxygen species, and inflammatory cytokines. In addition, down-regulation of epithelial cell markers such as E-cadherin facilitates de novo acquisition of mesenchymal phenotypes in MCs and production of extracellular matrices. Major mechanisms underlying the EMT of MCs include induction of oxidative stress, pro-inflammatory cytokines, endoplasmic reticulum stress and activation of the local renin-angiotensin system. Another mechanism of peritoneal EMT is mitigation of intrinsic defense mechanisms such as the peritoneal antioxidant system and anti-fibrotic peptide production in the peritoneal cavity. In addition to use of less bio-incompatible dialysates and optimum treatment of peritonitis in PD, therapies to prevent or alleviate peritoneal EMT have demonstrated a favorable effect on peritoneal function and structure, suggesting that EMT can be an early interventional target to preserve peritoneal integrity.
Collapse
Affiliation(s)
- Duk-Hee Kang
- Division of Nephrology, Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| |
Collapse
|
27
|
Conforti F, Ridley R, Brereton C, Alzetani A, Johnson B, Marshall BG, Fletcher SV, Ottensmeier CH, Richeldi L, Skipp P, Wang Y, Jones MG, Davies DE. Paracrine SPARC signaling dysregulates alveolar epithelial barrier integrity and function in lung fibrosis. Cell Death Discov 2020; 6:54. [PMID: 32637156 PMCID: PMC7327077 DOI: 10.1038/s41420-020-0289-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 06/08/2020] [Indexed: 12/23/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic scarring disease in which aging, environmental exposure(s) and genetic susceptibility have been implicated in disease pathogenesis, however, the causes and mechanisms of the progressive fibrotic cascade are still poorly understood. As epithelial-mesenchymal interactions are essential for normal wound healing, through human 2D and 3D in vitro studies, we tested the hypothesis that IPF fibroblasts (IPFFs) dysregulate alveolar epithelial homeostasis. Conditioned media from IPFFs exaggerated the wound-healing response of primary human Type II alveolar epithelial cells (AECs). Furthermore, AECs co-cultured with IPFFs exhibited irregular epithelialization compared with those co-cultured with control fibroblasts (NHLFs) or AECs alone, suggesting that epithelial homeostasis is dysregulated in IPF as a consequence of the abnormal secretory phenotype of IPFFs. Secretome analysis of IPFF conditioned media and functional studies identified the matricellular protein, SPARC, as a key mediator in the epithelial-mesenchymal paracrine signaling, with increased secretion of SPARC by IPFFs promoting persistent activation of alveolar epithelium via an integrin/focal adhesion/cellular-junction axis resulting in disruption of epithelial barrier integrity and increased macromolecular permeability. These findings suggest that in IPF fibroblast paracrine signaling promotes persistent alveolar epithelial activation, so preventing normal epithelial repair responses and restoration of tissue homeostasis. Furthermore, they identify SPARC-mediated paracrine signaling as a potential therapeutic target to promote the restoration of lung epithelial homoestasis in IPF patients.
Collapse
Affiliation(s)
- Franco Conforti
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
| | - Robert Ridley
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Christopher Brereton
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
| | - Aiman Alzetani
- Department of Thoracic Surgery, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Benjamin Johnson
- Cancer Sciences & NIHR and CRUK Experimental Cancer Sciences Unit, University of Southampton, Southampton, SO16 6YD UK
| | - Ben G. Marshall
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Sophie V. Fletcher
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Christian H. Ottensmeier
- University Hospital Southampton, Southampton, SO16 6YD UK
- Cancer Sciences & NIHR and CRUK Experimental Cancer Sciences Unit, University of Southampton, Southampton, SO16 6YD UK
| | - Luca Richeldi
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- Unità Operativa Complessa di Pneumologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Paul Skipp
- Centre for Proteomic Research, Institute for Life Sciences University of Southampton, Southampton, SO17 1BJ UK
| | - Yihua Wang
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ UK
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, SO17 1BJ UK
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- University Hospital Southampton, Southampton, SO16 6YD UK
| | - Donna E. Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ UK
| |
Collapse
|
28
|
Kelly D, Rothwell PM. Disentangling the multiple links between renal dysfunction and cerebrovascular disease. J Neurol Neurosurg Psychiatry 2020; 91:88-97. [PMID: 31511306 PMCID: PMC6952845 DOI: 10.1136/jnnp-2019-320526] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/29/2019] [Accepted: 08/07/2019] [Indexed: 11/30/2022]
Abstract
Chronic kidney disease (CKD) has a rapidly rising global prevalence, affecting as many as one-third of the population over the age of 75 years. CKD is a well-known risk factor for cardiovascular disease and, in particular, there is a strong association with stroke. Cohort studies and trials indicate that reduced glomerular filtration rate increases the risk of stroke by about 40% and that proteinuria increases the risk by about 70%. In addition, CKD is also strongly associated with subclinical cerebrovascular abnormalities, vascular cognitive impairment and dementia. The mechanisms responsible for these associations are currently unclear. CKD is associated with traditional risk factors such as hypertension, diabetes mellitus and atrial fibrillation, but non-traditional risk factors such as uraemia, oxidative stress, mineral and bone abnormalities, and dialysis-related factors, such as changes in cerebral blood flow or cardiac structure, are also postulated to play a role. Kidney disease can also impact and complicate the treatments used in acute stroke and in secondary prevention. In this review, we will outline our current understanding of the epidemiology and pathophysiology of cerebrovascular disease in CKD.
Collapse
Affiliation(s)
- Dearbhla Kelly
- Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Peter Malcolm Rothwell
- Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Claudin-7b and Claudin-h are required for controlling cilia morphogenesis in the zebrafish kidney. Mech Dev 2019; 161:103595. [PMID: 31887432 DOI: 10.1016/j.mod.2019.103595] [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] [Received: 09/04/2019] [Revised: 12/08/2019] [Accepted: 12/23/2019] [Indexed: 11/22/2022]
Abstract
Claudins are a family of proteins which are the most important components of the tight junctions. The location of Claudins on the renal tubule epithelial determines its paracellular transport characteristics, but whether Claudins have other functions in kidneys remains still unclear. Here, we showed that the transcripts encoding two Claudin family proteins, claudin-7b (cldn-7b) and claudin-h (cldn-h), were expressed in the transporting cells in the zebrafish pronephros. By knocking down of cldn-7b and cldn-h in zebrafish, we showed that these claudins morphants exhibited cystic kidneys accompanied with body curvature. Further analysis showed that down regulation of cldn-7b or cldn-h led to multiple defects in apico-basolateral polarity, cilia morphology and ciliary function in kidney. Moreover, the ciliary defect was confirmed by depletion of Cldn-7b or Cldn-h using CRISPR/Cas9 system. We also showed that both cldn-7b and cldn-h were genetically interacted with a well-known ciliary gene, arl13b. Deletion of arl13b led to curly cilia in the pronephros that phenocopied with cldn-7b and cldn-h morphants. Taken together, our data suggested that the tight junction protein, Cldn-7b and Cldn-h, regulate kidney development and function by affecting cilia morphology.
Collapse
|
30
|
Impact of Cold Ischemia Time on Outcomes of Deceased Donor Kidney Transplantation: An Analysis of a National Registry. Transplant Direct 2019; 5:e448. [PMID: 31165083 PMCID: PMC6511440 DOI: 10.1097/txd.0000000000000888] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/21/2019] [Accepted: 02/24/2019] [Indexed: 11/25/2022] Open
Abstract
Background Cold ischemia time (CIT) is known to impact kidney graft survival rates. We compare the impact of CIT on graft failure and mortality in circulatory death versus brain death donor kidneys and how it relates to donor age. Methods We used the prospective Dutch Organ Transplantation Registry to include 2153 adult recipients of brain death (n = 1266) and circulatory death (n = 887) donor kidneys after static cold storage from transplants performed between 2005 and 2012. CIT was modeled nonlinearly with splines. Associations and interactions between CIT, donor type, donor age, 5-year (death-censored) graft survival, and mortality were evaluated. Results The median CIT was 16.2 hours (interquartile range 12.8-20), ranging from 3.4 to 44.7 hours for brain death and 4.7 to 46.6 hours for circulatory death donor kidneys. At >12 hours of CIT, we observed an increased risk of graft failure in kidneys donated after circulatory death versus after brain death. This risk rose significantly at >22 hours of CIT (hazard ratio 1.45; 95% confidence interval, 1.01-2.49; P = 0.043). Kidneys that came from 60-year-old circulatory death donors demonstrated elevated hazard risk at 19 hours of CIT, a shorter timeline than that for kidneys that came from brain death donors of the same age (hazard ratio 1.33; 95% confidence interval, 1.00-1.78; P = 0.045). The additional harmful effects of increased CIT in kidneys from circulatory-death donors were also found for death-censored graft failure but did not affect mortality rates in any significant way. Conclusions The findings support the hypothesis that prolonged cold ischemia is more harmful for circulatory death donor kidneys that have already been subjected to a permissible period of warm ischemia. Efforts should be made to reduce CIT, especially for older circulatory death donor kidneys.
Collapse
|
31
|
You ZP, Chen SS, Yang ZY, Li SR, Xiong F, Liu T, Fu SH. GEP100/ARF6 regulates VEGFR2 signaling to facilitate high-glucose-induced epithelial-mesenchymal transition and cell permeability in retinal pigment epithelial cells. Am J Physiol Cell Physiol 2018; 316:C782-C791. [PMID: 30540496 DOI: 10.1152/ajpcell.00312.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell permeability and epithelial-mesenchymal transition (EMT) were found to be enhanced in diabetic retinopathy, and the aim of this study was to investigate the underlying mechanism. ARPE-19 cell line or primary retinal pigment epithelial (RPE) cells were cultured under high or normal glucose conditions. Specific shRNAs were employed to knock down ADP-ribosylation factor 6 (ARF6), GEP100, or VEGF receptor 2 (VEGFR2) in ARPE-19 or primary RPE cells. Cell migration ability was measured using Transwell assay. Western blotting was used to measure indicated protein levels. RPE cells treated with high glucose showed increased cell migration, paracellular permeability, EMT, and expression of VEGF. Knockdown of VEGFR2 inhibited the high-glucose-induced effects on RPE cells via inactivation of ARF6 and MAPK pathways. Knockdown ARF6 or GEP100 led to inhibition of high-glucose-induced effects via inactivation of VEGFR2 pathway. Knockdown of ARF6, but not GEP100, decreased high-glucose-induced internalization of VEGFR2. High-glucose enhances EMT and cell permeability of RPE cells through activation of VEGFR2 and ARF6/GEP100 pathways, which form a positive feedback loop to maximize the activation of VEGF/VEGFR2 signaling.
Collapse
Affiliation(s)
- Zhi-Peng You
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Shan-Shan Chen
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Zhong-Yi Yang
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Shu-Rong Li
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Fan Xiong
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Ting Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Shu-Hua Fu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| |
Collapse
|
32
|
Kim DY, Woo YM, Lee S, Oh S, Shin Y, Shin JO, Park EY, Ko JY, Lee EJ, Bok J, Yoo KH, Park JH. Impact of miR-192 and miR-194 on cyst enlargement through EMT in autosomal dominant polycystic kidney disease. FASEB J 2018; 33:2870-2884. [PMID: 30332302 DOI: 10.1096/fj.201800563rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Altered miRNA (miR) expression occurs in various diseases. However, the therapeutic effect of miRNAs in autosomal dominant polycystic kidney disease (ADPKD) is unclear. Genome-wide analyses of miRNA expression and DNA methylation status were conducted to identify crucial miRNAs in end-stage ADPKD. miR-192 and -194 levels were down-regulated with hypermethylation at these loci, mainly in the intermediate and late stages, not in the early stage, of cystogenesis, suggesting their potential impact on cyst expansion. Cyst expansion has been strongly associated with endothelial-mesenchymal transition (EMT). Zinc finger E-box-binding homeobox-2 and cadherin-2, which are involved in EMT, were directly regulated by miR-192 and -194. The therapeutic effect of miR-192 and -194 in vivo and in vitro were assessed. Restoring these miRs by injection of precursors influenced the reduced size of cysts in Pkd1 conditional knockout mice. miR-192 and -194 may act as potential therapeutic targets to control the expansion and progression of cysts in patients with ADPKD.-Kim, D. Y., Woo, Y. M., Lee, S., Oh, S., Shin, Y., Shin, J.-O., Park, E. Y., Ko, J. Y., Lee, E. J., Bok, J., Yoo, K. H., Park, J. H. Impact of miR-192 and miR-194 on cyst enlargement through EMT in autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Do Yeon Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Yu Mi Woo
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Sunyoung Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Sumin Oh
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Yubin Shin
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Jeong-Oh Shin
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea; and
| | - Eun Young Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Je Yeong Ko
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Eun Ji Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea; and.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyung Hyun Yoo
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| | - Jong Hoon Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea.,Research Institute of Women's Health, Sookmyung Women's University, Seoul, South Korea
| |
Collapse
|
33
|
Song LJ, Zhou LL, Wang M, Liu F, Xiong L, Xiang F, Yu F, He XL, Xu JJ, Shi HZ, Xin JB, Ye H, Ma WL. Lethal (2) giant larvae regulates pleural mesothelial cell polarity in pleural fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1201-1210. [PMID: 29842893 DOI: 10.1016/j.bbamcr.2018.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/20/2018] [Accepted: 05/24/2018] [Indexed: 12/29/2022]
Abstract
Pleural fibrosis is barely reversible and the underlying mechanisms are poorly understood. Pleural mesothelial cells (PMCs) which have apical-basal polarity play a key role in pleural fibrosis. Loss of cell polarity is involved in the development of fibrotic diseases. Partition defective protein (PAR) complex is a key regulator of cell polarity. However, changes of PMC polarity and PAR complex in pleural fibrosis are still unknown. In this study, we observed that PMC polarity was lost in fibrotic pleura. Next we found increased Lethal (2) giant larvae (Lgl) bound with aPKC and PAR-6B competing against PAR-3A in PAR complex, which led to cell polarity loss. Then we demonstrated that Lgl1 siRNA prevented cell polarity loss in PMCs, and Lgl1 conditional knockout (ER-Cre+/-Lgl1flox/flox) attenuated pleural fibrosis in a mouse model. Our data indicated that Lgl1 regulates cell polarity of PMCs, inhibition of Lgl1 and maintenance of cell polarity in PMCs could be a potential therapeutic treatment approach for pleural fibrosis.
Collapse
Affiliation(s)
- Lin-Jie Song
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li-Ling Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fei Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China
| | - Fei Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China
| | - Fan Yu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China
| | - Xin-Liang He
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China
| | - Juan-Juan Xu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China
| | - Huan-Zhong Shi
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jian-Bao Xin
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China
| | - Hong Ye
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China.
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases, Ministry of Health of China, Wuhan 430030, China.
| |
Collapse
|
34
|
Sheen YH, Jee HM, Kim DH, Ha EK, Jeong IJ, Lee SJ, Baek HS, Lee SW, Lee K, Lee KS, Jung Y, Sung M, Kim MA, Han MY. Serum zonulin is associated with presence and severity of atopic dermatitis in children, independent of total IgE and eosinophil. Clin Exp Allergy 2018; 48:1059-1062. [DOI: 10.1111/cea.13158] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Y. H. Sheen
- Department of Pediatrics CHA Gangnam Medical Center CHA University Seoul Korea
| | - H. M. Jee
- Department of Pediatrics CHA Bundang Medical Center CHA University Seongnam Korea
| | - D. H. Kim
- Department of Dermatology CHA Bundang Medical Center CHA University Seongnam Korea
| | - E. K. Ha
- Department of Pediatrics CHA Bundang Medical Center CHA University Seongnam Korea
| | - I. J. Jeong
- Department of Dermatology CHA Bundang Medical Center CHA University Seongnam Korea
| | - S. J. Lee
- Department of Pediatrics CHA Bundang Medical Center CHA University Seongnam Korea
| | - H. S. Baek
- Department of Pediatrics Hallym University Kangdong Sacred Heart Hospital Hallym University College of Medicine Seoul Korea
| | | | - K.‐J. Lee
- Department of Information and Statistics Korea National Open University Seoul Korea
| | - K. S. Lee
- Department of Pediatrics CHA Bundang Medical Center CHA University Seongnam Korea
| | - Y.‐H. Jung
- Department of Pediatrics CHA Bundang Medical Center CHA University Seongnam Korea
| | - M. Sung
- Department of Pediatrics Inje University Haeundae Paik Hospital Busan Korea
| | - M. A. Kim
- Department of Internal Medicine CHA Bundang Medical Center CHA University Seongnam Korea
| | - M. Y. Han
- Department of Pediatrics CHA Bundang Medical Center CHA University Seongnam Korea
| |
Collapse
|
35
|
De Rosa S, Antonelli M, Ronco C. Hypothermia and kidney: a focus on ischaemia-reperfusion injury. Nephrol Dial Transplant 2018; 32:241-247. [PMID: 28186567 DOI: 10.1093/ndt/gfw038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/03/2016] [Indexed: 01/01/2023] Open
Abstract
Cellular damage after reperfusion of ischaemic tissue is defined as ischaemia–reperfusion injury (IRI). Hypothermia is able to decrease oxygen consumption, preventing a rapid loss of mitochondrial activity. However, even though cooling can help to decrease the deleterious effects of ischaemia, the consequences are not exclusively beneficial, such that hypothermic storage is a compromise between benefits and harm. The present review details the relationship between renal IRI and hypothermia, describing the pathophysiology of IRI and hypothermic protection through experimental evidence. Although experimental models of renal IRI are a valuable tool for understanding the pathophysiology of renal ischaemia–reperfusion, the clinical transfer of experimental results has several limitations, particularly because of anatomical and physiological differences. In this review limitations of animal models but also hypothermia as a strategy to protect the kidney from IRI are discussed. We also attempt to describe three clinical scenarios where hypothermia is used in clinical settings of IRI: transplantation, deceased donors and post-cardiac arrest.
Collapse
Affiliation(s)
- Silvia De Rosa
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy.,Department of Anaesthesia and Intensive Care, Catholic University, Rome, Italy.,Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Vicenza, Italy
| | - Massimo Antonelli
- Department of Anaesthesia and Intensive Care, Catholic University, Rome, Italy
| | - Claudio Ronco
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy.,Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Vicenza, Italy
| |
Collapse
|
36
|
Mongelli-Sabino BM, Canuto LP, Collares-Buzato CB. Acute and chronic exposure to high levels of glucose modulates tight junction-associated epithelial barrier function in a renal tubular cell line. Life Sci 2017; 188:149-157. [PMID: 28882647 DOI: 10.1016/j.lfs.2017.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 09/01/2017] [Accepted: 09/02/2017] [Indexed: 01/13/2023]
Abstract
AIMS Type 2 diabetes mellitus (T2DM) is one of the most prevalent diseases worldwide. Diabetic nephropathy (DN) is a complication of diabetes and the mechanisms underlying onset and progression of this disease are not fully understood. It has been shown that hyperglycemia is an independent factor to predict the development of DN in individuals with T2DM, however, a link between high plasma glucose levels and renal tubular injuries in DN remains unknown. In this study, we investigated the effect of high levels of glucose (i.e. 180 or 360mg/dL) for up to 24h (acute) or over 72h (chronic) upon tight junction (TJ)-mediated epithelial barrier integrity of the kidney tubular cell line, MDCK. METHODS/KEY FINDINGS High levels of glucose (180 and 360mg/dL) induced a decrease in transepithelial electrical resistance associated with an increase in TJ cation selectivity at 24h or in TJ permeability to a paracellular marker, Lucifer Yellow, at 72h-exposure when compared to control group (exposed to 100mg/dL glucose). Immunofluorescence analyses showed that glucose treatment induced a significant decrease in the tight junctional content of claudins-1 and -3 as well as a significant increase in claudin-2 (particularly at 24h-exposure) and a time-dependent change in occludin/ZO-1 junctional content. The analyses of total cell content of these junctional proteins by Western blot did not reveal significant changes, except in claudin-2 expression. SIGNIFICANCE Our data suggest that high levels of glucose induce time-dependence changes in TJ structure in MDCK monolayers, suggesting a possible link between hyperglycemia-induced tubular epithelial barrier disruption and diabetic nephropathy.
Collapse
Affiliation(s)
- B M Mongelli-Sabino
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - L P Canuto
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - C B Collares-Buzato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
37
|
Marciano DK. A holey pursuit: lumen formation in the developing kidney. Pediatr Nephrol 2017; 32:7-20. [PMID: 26902755 PMCID: PMC5495142 DOI: 10.1007/s00467-016-3326-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/23/2015] [Accepted: 01/12/2016] [Indexed: 02/07/2023]
Abstract
The formation of polarized epithelial tubules is a hallmark of kidney development. One of the fundamental principles in tubulogenesis is that epithelia coordinate the polarity of individual cells with the surrounding cells and matrix. A central feature in this process is the segregation of membranes into spatially and functionally distinct apical and basolateral domains, and the generation of a luminal space at the apical surface. This review examines our current understanding of the cellular and molecular mechanisms that underlie the establishment of apical-basal polarity and lumen formation in developing renal epithelia, including the roles of cell-cell and cell-matrix interactions and polarity complexes. We highlight growing evidence from animal models, and correlate these findings with models of tubulogenesis from other organ systems, and from in vitro studies.
Collapse
Affiliation(s)
- Denise K. Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. H5.102, Dallas, TX 75390-8856
| |
Collapse
|
38
|
Histamine type 1-receptor activation by low dose of histamine undermines human glomerular slit diaphragm integrity. Pharmacol Res 2016; 114:27-38. [DOI: 10.1016/j.phrs.2016.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 11/20/2022]
|
39
|
Kim CS, Shin DM. Improper hydration induces global gene expression changes associated with renal development in infant mice. GENES AND NUTRITION 2016; 11:28. [PMID: 27785155 PMCID: PMC5072351 DOI: 10.1186/s12263-016-0544-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/03/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND The kidney is a major organ in which fluid balance and waste excretion is regulated. For the kidney to achieve maturity with functions, normal renal developmental processes need to occur. Comprehensive genetic programs underlying renal development during the prenatal period have been widely studied. However, postnatal renal development, from infancy to the juvenile period, has not been studied yet. Here, we investigated whether structural and functional kidney development was still ongoing in early life by analyzing the renal transcriptional networks of infant (4 weeks old) and juvenile (7 weeks old) mice. We further examined the effects of dehydration on kidney development to unravel the mechanistic bases underlying deteriorative impact of pediatric dehydration on renal development. METHODS 3-week-old infant mice that just finished weaning period were provided limited access to a water for fifteen minutes per day for one week (RES 1W) and four weeks (RES 4W) to induce dehydration while control group consumed water ad libitum with free access to the water bottle. Transcriptome analysis was conducted to understand physiological changes during postnatal renal development and dehydration. RESULTS Kidneys in 4-week- and 7-week-old mice showed significantly distinctive functional gene networks. Gene sets related to cell cycle regulators, fetal kidney patterning molecules, and immature basement membrane integrity were upregulated in infantile kidneys while heightened expressions of genes associated with ion transport and drug metabolism were observed in juvenile kidneys. Dehydration during infancy suppressed renal growth by interrupting the SHH signaling pathway, which targets cell cycle regulators. Importantly, it is likely that disruption of the developmental program ultimately led to a decline in gene expression associated with basement membrane integrity. CONCLUSIONS Altogether, we demonstrate transcriptional events during renal development in infancy and show that the impacts of inadequate water intake in the early postnatal state heavily rely on the impairment of normal renal development. Here, we provide a meaningful perspective of renal development in infancy with a molecular and physiological explanation of why infants are more vulnerable to dehydration than adults. These results provide new insights into the molecular effects of dehydration on renal physiology and indicate that optimal nutritional interventions are necessary for pediatric renal development.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, Seoul National University, Seoul, 08826 South Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University, Seoul, 08826 South Korea ; Research Institute of Human Ecology, Seoul National University, Seoul, 08826 South Korea
| |
Collapse
|
40
|
Lau WL, Huisa BN, Fisher M. The Cerebrovascular-Chronic Kidney Disease Connection: Perspectives and Mechanisms. Transl Stroke Res 2016; 8:67-76. [PMID: 27628245 PMCID: PMC5241336 DOI: 10.1007/s12975-016-0499-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/21/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is an independent risk factor for the development of cerebrovascular disease, particularly small vessel disease which can manifest in a variety of phenotypes ranging from lacunes to microbleeds. Small vessel disease likely contributes to cognitive dysfunction in the CKD population. Non-traditional risk factors for vascular injury in uremia include loss of calcification inhibitors, hyperphosphatemia, increased blood pressure variability, elastinolysis, platelet dysfunction, and chronic inflammation. In this review, we discuss the putative pathways by which these mechanisms may promote cerebrovascular disease and thus increase risk of future stroke in CKD patients.
Collapse
Affiliation(s)
- Wei Ling Lau
- Department of Medicine, Division of Nephrology, University of California, Irvine, CA, USA
| | - Branko N Huisa
- Department of Neurology, University of California, San Diego, CA, USA
| | - Mark Fisher
- Departments of Neurology, Anatomy & Neurobiology, and Pathology & Laboratory Medicine, University of California, Irvine, CA, USA. .,Department of Neurology, UC Irvine Medical Center, 101 The City Drive South, Shanbrom Hall, Room 121, Orange, CA, 92868, USA.
| |
Collapse
|
41
|
Martovetsky G, Bush KT, Nigam SK. Kidney versus Liver Specification of SLC and ABC Drug Transporters, Tight Junction Molecules, and Biomarkers. ACTA ACUST UNITED AC 2016; 44:1050-60. [PMID: 27044799 DOI: 10.1124/dmd.115.068254] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/30/2016] [Indexed: 01/04/2023]
Abstract
The hepatocyte nuclear factors, Hnf1a and Hnf4a, in addition to playing key roles in determining hepatocyte fate, have been implicated as candidate lineage-determining transcription factors in the kidney proximal tubule (PT) [Martovetsky et. al., (2012) Mol Pharmacol 84:808], implying an additional level of regulation that is potentially important in developmental and/or tissue-engineering contexts. Mouse embryonic fibroblasts (MEFs) transduced with Hnf1a and Hnf4a form tight junctions and express multiple PT drug transporters (e.g., Slc22a6/Oat1, Slc47a1/Mate1, Slc22a12/Urat1, Abcg2/Bcrp, Abcc2/Mrp2, Abcc4/Mrp4), nutrient transporters (e.g., Slc34a1/NaPi-2, Slco1a6), and tight junction proteins (occludin, claudin 6, ZO-1/Tjp1, ZO-2/Tjp2). In contrast, the coexpression (with Hnf1a and Hnf4a) of GATA binding protein 4 (Gata4), as well as the forkhead box transcription factors, Foxa2 and Foxa3, in MEFs not only downregulates PT markers but also leads to upregulation of several hepatocyte markers, including albumin, apolipoprotein, and transferrin. A similar result was obtained with primary mouse PT cells. Thus, the presence of Gata4 and Foxa2/Foxa3 appears to alter the effect of Hnf1a and Hnf4a by an as-yet unidentified mechanism, leading toward the generation of more hepatocyte-like cells as opposed to cells exhibiting PT characteristics. The different roles of Hnf4a in the kidney and liver was further supported by reanalysis of ChIP-seq data, which revealed Hnf4a colocalization in the kidney near PT-enriched genes compared with those genes enriched in the liver. These findings provide valuable insight, not only into the developmental, and perhaps organotypic, regulation of drug transporters, drug-metabolizing enzymes, and tight junctions, but also for regenerative medicine strategies aimed at restoring the function of the liver and/or kidney (acute kidney injury, AKI; chronic kidney disease, CKD).
Collapse
Affiliation(s)
- Gleb Martovetsky
- Department of Pediatrics (G.M., K.T.B., S.K.N.), Department of Medicine, Division of Nephrology and Hypertension, (S.K.N.), and Department of Cellular and Molecular Medicine (S.K.N.), University of California, San Diego, La Jolla, California
| | - Kevin T Bush
- Department of Pediatrics (G.M., K.T.B., S.K.N.), Department of Medicine, Division of Nephrology and Hypertension, (S.K.N.), and Department of Cellular and Molecular Medicine (S.K.N.), University of California, San Diego, La Jolla, California
| | - Sanjay K Nigam
- Department of Pediatrics (G.M., K.T.B., S.K.N.), Department of Medicine, Division of Nephrology and Hypertension, (S.K.N.), and Department of Cellular and Molecular Medicine (S.K.N.), University of California, San Diego, La Jolla, California
| |
Collapse
|
42
|
Liu Y, Dong Z, Liu H, Zhu J, Liu F, Chen G. Transition of mesothelial cell to fibroblast in peritoneal dialysis: EMT, stem cell or bystander? Perit Dial Int 2015; 35:14-25. [PMID: 25700459 DOI: 10.3747/pdi.2014.00188] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Long-term peritoneal dialysis (PD) can lead to fibrotic changes in the peritoneum, characterized by loss of mesothelial cells (MCs) and thickening of the submesothelial area with an accumulation of collagen and myofibroblasts. The origin of myofibroblasts is a central question in peritoneal fibrosis that remains unanswered at present. Numerous clinical and experimental studies have suggested that MCs, through epithelial-mesenchymal transition (EMT), contribute to the pool of peritoneal myofibroblasts. However, recent work has placed significant doubts on the paradigm of EMT in organ fibrogenesis (in the kidney particularly), highlighting the need to reconsider the role of EMT in the generation of myofibroblasts in peritoneal fibrosis. In particular, selective cell isolation and lineage-tracing experiments have suggested the existence of progenitor cells in the peritoneum, which are able to switch to fibroblast-like cells when stimulated by the local environment. These findings highlight the plastic nature of MCs and its contribution to peritoneal fibrogenesis. In this review, we summarize the key findings and caveats of EMT in organ fibrogenesis, with a focus on PD-related peritoneal fibrosis, and discuss the potential of peritoneal MCs as a source of myofibroblasts.
Collapse
Affiliation(s)
- Yu Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Hong Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Jiefu Zhu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Fuyou Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Guochun Chen
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
43
|
Hascoet P, Chesnel F, Le Goff C, Le Goff X, Arlot-Bonnemains Y. Unconventional Functions of Mitotic Kinases in Kidney Tumorigenesis. Front Oncol 2015; 5:241. [PMID: 26579493 PMCID: PMC4621426 DOI: 10.3389/fonc.2015.00241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/12/2015] [Indexed: 01/25/2023] Open
Abstract
Human tumors exhibit a variety of genetic alterations, including point mutations, translocations, gene amplifications and deletions, as well as aneuploid chromosome numbers. For carcinomas, aneuploidy is associated with poor patient outcome for a large variety of tumor types, including breast, colon, and renal cell carcinoma. The Renal cell carcinoma (RCC) is a heterogeneous carcinoma consisting of different histologic types. The clear renal cell carcinoma (ccRCC) is the most common subtype and represents 85% of the RCC. Central to the biology of the ccRCC is the loss of function of the Von Hippel–Lindau gene, but is also associated with genetic instability that could be caused by abrogation of the cell cycle mitotic spindle checkpoint and may involve the Aurora kinases, which regulate centrosome maturation. Aneuploidy can also result from the loss of cell–cell adhesion and apical–basal cell polarity that also may be regulated by the mitotic kinases (polo-like kinase 1, casein kinase 2, doublecortin-like kinase 1, and Aurora kinases). In this review, we describe the “non-mitotic” unconventional functions of these kinases in renal tumorigenesis.
Collapse
Affiliation(s)
- Pauline Hascoet
- UMR 6290 (IGDR), CNRS, University Rennes-1 , Rennes , France
| | - Franck Chesnel
- UMR 6290 (IGDR), CNRS, University Rennes-1 , Rennes , France
| | - Cathy Le Goff
- UMR 6290 (IGDR), CNRS, University Rennes-1 , Rennes , France
| | - Xavier Le Goff
- UMR 6290 (IGDR), CNRS, University Rennes-1 , Rennes , France
| | | |
Collapse
|
44
|
Claverie-Martin F. Familial hypomagnesaemia with hypercalciuria and nephrocalcinosis: clinical and molecular characteristics. Clin Kidney J 2015; 8:656-64. [PMID: 26613020 PMCID: PMC4655790 DOI: 10.1093/ckj/sfv081] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/04/2015] [Indexed: 01/19/2023] Open
Abstract
Familial hypomagnesaemia with hypercalciuria and nephrocalcinosis (FHHNC) is an autosomal-recessive renal tubular disorder characterized by excessive urinary losses of magnesium and calcium, bilateral nephrocalcinosis and progressive chronic renal failure. Presentation with FHHNC symptoms generally occurs early in childhood or before adolescence. At present, the only therapeutic option is supportive and consists of oral magnesium supplementation and thiazide diuretics. However, neither treatment seems to have a significant effect on the levels of serum magnesium or urine calcium or on the decline of renal function. In end-stage renal disease patients, renal transplantation is the only effective approach. This rare disease is caused by mutations in the CLDN16 or CLDN19 genes. Patients with mutations in CLDN19 also present severe ocular abnormalities such as myopia, nystagmus and macular colobamata. CLDN16 and CLDN19 encode the tight-junction proteins claudin-16 and claudin-19, respectively, which are expressed in the thick ascending limb of Henle's loop and form an essential complex for the paracellular reabsorption of magnesium and calcium. Claudin-19 is also expressed in retinal epithelium and peripheral neurons. Research studies using mouse and cell models have generated significant advances on the understanding of the pathophysiology of FHHNC. A recent finding has established that another member of the claudin family, claudin-14, plays a key regulatory role in paracellular cation reabsorption by inhibiting the claudin-16-claudin-19 complex. Furthermore, several studies on the molecular and cellular consequences of disease-causing CLDN16 and CLDN19 mutations have provided critical information for the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Felix Claverie-Martin
- Unidad de Investigación, Hospital Nuestra Señora de Candelaria , Santa Cruz de Tenerife , Spain
| |
Collapse
|
45
|
Guo X, Wang M, Zhao Y, Wang X, Shen M, Zhu F, Shi C, Xu M, Li X, Peng F, Zhang H, Feng Y, Xie Y, Xu X, Jia W, He R, Jiang J, Hu J, Tian R, Qin R. Par3 regulates invasion of pancreatic cancer cells via interaction with Tiam1. Clin Exp Med 2015; 16:357-65. [PMID: 26084985 DOI: 10.1007/s10238-015-0365-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/03/2015] [Indexed: 12/16/2022]
Abstract
The conserved polarity complex, which comprises partitioning-defective proteins Par3, Par6, and the atypical protein kinase C, affects various cell-polarization events, including assembly of tight junctions. Control of tight junction assembly is closely related to invasion and migration potential. However, as the importance of conserved polarity complexes in regulating pancreatic cancer invasion and metastasis is unclear, we investigated their role and mechanism in pancreatic cancers. We first detect that the key protein of the conserved polarity complex finds that only Par3 is down-regulated in pancreatic cancer tissues while Par6 and aPKC show no difference. What is more, Par3 tissues level was significantly and positively associated with patient overall survival. Knocking-down Par3 promotes pancreatic cancer cells invasion and migration. And Par3 requires interaction with Tiam1 to affect tight junction assembly, and then affect invasion and migration of pancreatic cancer cells. Then, we find that tight junction marker protein ZO-1 and claudin-1 are down-regulated in pancreatic cancer tissues. And the relationship of the expression of Par3 and ZO-1 in pancreatic cancer tissue is linear correlation. We establish liver metastasis model of human pancreatic cancer cells in Balb/c nude mice and find that knocking down Par3 promotes invasion and metastasis and disturbs tight junction assembly in vivo. Taken together, these results suggest that the Par3 regulates invasion and metastasis in pancreatic cancers by controlling tight junction assembly.
Collapse
Affiliation(s)
- Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Yan Zhao
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Xin Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Ming Shen
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Feng Zhu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Chengjian Shi
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Meng Xu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Xu Li
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Feng Peng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Hang Zhang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Yechen Feng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Yu Xie
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Xiaodong Xu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Wei Jia
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Jianxin Jiang
- Department of Hepatic-Biliary-Pancreatic Surgery, Hubei Cancer Hospital, Wuhan City, People's Republic of China
| | - Jun Hu
- Department of Colon Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Rui Tian
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China.
| |
Collapse
|
46
|
Kang MK, Park SH, Choi YJ, Shin D, Kang YH. Chrysin inhibits diabetic renal tubulointerstitial fibrosis through blocking epithelial to mesenchymal transition. J Mol Med (Berl) 2015; 93:759-72. [PMID: 26062793 DOI: 10.1007/s00109-015-1301-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 05/03/2015] [Accepted: 05/12/2015] [Indexed: 01/28/2023]
Abstract
UNLABELLED Renal fibrosis is a crucial event in the pathogenesis of diabetic nephropathy (DN). The process known as epithelial to mesenchymal transition (EMT) contributes to the accumulation of matrix proteins in kidneys, in which renal tubular epithelial cells play an important role in progressive renal fibrosis. The current study investigated that chrysin (5,7-dihydroxyflavone) present in bee propolis and herbs, inhibited renal tubular EMT and tubulointerstitial fibrosis due to chronic hyperglycemia. Human renal proximal tubular epithelial cells (RPTEC) were incubated in media containing 5.5 mM glucose, 27.5 mM mannitol (as an osmotic control), or 33 mM glucose (HG) in the absence and presence of 1-20 μM chrysin for 72 h. Chrysin significantly inhibited high glucose-induced renal EMT through blocking expression of the mesenchymal markers vimentin, α-smooth muscle actin, and fibroblast-specific protein-1 in RPTEC and db/db mice. Chrysin reversed the HG-induced down-regulation of the epithelial marker E-cadherin and the HG-enhanced N-cadherin induction in RPTEC. In addition, chrysin inhibited the production of collagen IV in tubular cells and the deposition of collagen fibers in mouse kidneys. Furthermore, chrysin blocked tubular cell migration concurrent with decreasing matrix metalloproteinase-2 activity, indicating epithelial cell derangement and tubular basement membrane disruption. Chrysin restored the induction of the tight junction proteins Zona occludens protein-1 (ZO-1) and occludin downregulated in diabetic mice. Chrysin inhibited renal tubular EMT-mediated tubulointerstitial fibrosis caused by chronic hyperglycemia. Therefore, chrysin may be a potent renoprotective agent for the treatment of renal fibrosis-associated DN. KEY MESSAGES • Glucose increases renal tubular epithelial induction of vimentin, α-SMA and FSP-1. • Glucose enhances renal EMT by blocking tubular epithelial E-cadherin expression. • Chrysin inhibits tubular EMT-mediated tubulointerstitial fibrosis in mouse kidneys. • Chrysin restores renal tubular induction of ZO-1 and occludin downregulated in diabetic mice. • Chrysin blocks glucose-induced renal tubular cell migration with reducing MMP-2 activity.
Collapse
Affiliation(s)
- Min-Kyung Kang
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do, 200-702, Republic of Korea
| | | | | | | | | |
Collapse
|
47
|
Qiu X, Fu K, Zhao X, Zhang Y, Yuan Y, Zhang S, Gu X, Guo H. Protective effects of astaxanthin against ischemia/reperfusion induced renal injury in mice. J Transl Med 2015; 13:28. [PMID: 25623758 PMCID: PMC4323259 DOI: 10.1186/s12967-015-0388-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/12/2015] [Indexed: 12/11/2022] Open
Abstract
Astaxanthin (ATX) is a powerful antioxidant that occurs naturally in a wide variety of living organisms. Previous studies have shown that ATX has effects of eliminating oxygen free radicals and can protect organs from ischemia/reperfusion (IR) induced injury. The present study was designed to further investigate the protective effects of ATX on oxidative stress induced toxicity in tubular epithelial cells and on IR induced renal injury in mice. ATX, at a concentration of 250 nM, attenuated 100 μM H2O2-inudced viability decrease of tubular epithelial cells. In vivo, ATX preserved renal function 12 h or 24 h post IR. Pretreatment of ATX via oral gavage for 14 consecutive days prior to IR dramatically prevented IR induced histological damage 24 h post IR. Histological results showed that the pathohistological score, number of apoptotic cells, and the expression of α-smooth muscle actin were significantly decreased by pretreatment of ATX. In addition, oxidative stress and inflammation in kidney samples were significantly reduced by ATX 24 h post IR. Taken together, the current study suggests that pretreatment of ATX is effective in preserving renal function and histology via antioxidant activity.
Collapse
Affiliation(s)
- Xuefeng Qiu
- Department of Urology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China. .,Institute of Urology, Nanjing University, Nanjing, 210093, China. .,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093, China.
| | - Kai Fu
- Department of Urology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China. .,Institute of Urology, Nanjing University, Nanjing, 210093, China.
| | - Xiaozhi Zhao
- Department of Urology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China. .,Institute of Urology, Nanjing University, Nanjing, 210093, China.
| | - Yanting Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093, China.
| | - Yimin Yuan
- Department of Urology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China.
| | - Shiwei Zhang
- Department of Urology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China.
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
48
|
Simone S, Rascio F, Castellano G, Divella C, Chieti A, Ditonno P, Battaglia M, Crovace A, Staffieri F, Oortwijn B, Stallone G, Gesualdo L, Pertosa G, Grandaliano G. Complement-dependent NADPH oxidase enzyme activation in renal ischemia/reperfusion injury. Free Radic Biol Med 2014; 74:263-73. [PMID: 25017967 DOI: 10.1016/j.freeradbiomed.2014.07.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 01/06/2023]
Abstract
NADPH oxidase plays a central role in mediating oxidative stress during heart, liver, and lung ischemia/reperfusion injury, but limited information is available about NADPH oxidase in renal ischemia/reperfusion injury. Our aim was to investigate the activation of NADPH oxidase in a swine model of renal ischemia/reperfusion damage. We induced renal ischemia/reperfusion in 10 pigs, treating 5 of them with human recombinant C1 inhibitor, and we collected kidney biopsies before ischemia and 15, 30, and 60 min after reperfusion. Ischemia/reperfusion induced a significant increase in NADPH oxidase 4 (NOX-4) expression at the tubular level, an upregulation of NOX-2 expression in infiltrating monocytes and myeloid dendritic cells, and 8-oxo-7,8-dihydro-2'-deoxyguanosine synthesis along with a marked upregulation of NADPH-dependent superoxide generation. This burden of oxidative stress was associated with an increase in tubular and interstitial expression of the myofibroblast marker α-smooth muscle actin (α-SMA). Interestingly, NOX-4 and NOX-2 expression and the overall NADPH oxidase activity as well as α-SMA expression and 8-oxo-7,8-dihydro-2'-deoxyguanosine synthesis were strongly reduced in C1-inhibitor-treated animals. In vitro, when we incubated tubular cells with the anaphylotoxin C3a, we observed an enhanced NADPH oxidase activity and α-SMA protein expression, which were both abolished by NOX-4 silencing. In conclusion, our findings suggest that NADPH oxidase is activated during ischemia/reperfusion in a complement-dependent manner and may play a potential role in the pathogenesis of progressive renal damage in this setting.
Collapse
Affiliation(s)
- S Simone
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - F Rascio
- Nephrology, Dialysis, and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy.
| | - G Castellano
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - C Divella
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - A Chieti
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - P Ditonno
- Urology, Andrology, and Renal Transplantation Unit, and Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - M Battaglia
- Urology, Andrology, and Renal Transplantation Unit, and Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - A Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - F Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | | | - G Stallone
- Nephrology, Dialysis, and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
| | - L Gesualdo
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - G Pertosa
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro," 70121 Bari, Italy
| | - G Grandaliano
- Nephrology, Dialysis, and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
| |
Collapse
|
49
|
Kyuno D, Yamaguchi H, Ito T, Kono T, Kimura Y, Imamura M, Konno T, Hirata K, Sawada N, Kojima T. Targeting tight junctions during epithelial to mesenchymal transition in human pancreatic cancer. World J Gastroenterol 2014; 20:10813-10824. [PMID: 25152584 PMCID: PMC4138461 DOI: 10.3748/wjg.v20.i31.10813] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 05/05/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer continues to be a leading cause of cancer-related death worldwide and there is an urgent need to develop novel diagnostic and therapeutic strategies to reduce the mortality of patients with this disease. In pancreatic cancer, some tight junction proteins, including claudins, are abnormally regulated and therefore are promising molecular targets for diagnosis, prognosis and therapy. Claudin-4 and -18 are overexpressed in human pancreatic cancer and its precursor lesions. Claudin-4 is a high affinity receptor of Clostridium perfringens enterotoxin (CPE). The cytotoxic effects of CPE and monoclonal antibodies against claudin-4 are useful as novel therapeutic tools for pancreatic cancer. Claudin-18 could be a putative marker and therapeutic target with prognostic implications for patients with pancreatic cancer. Claudin-1, -7, tricellulin and marvelD3 are involved in epithelial to mesenchymal transition (EMT) of pancreatic cancer cells and thus might be useful as biomarkers during disease. Protein kinase C is closely related to EMT of pancreatic cancer and regulates tight junctions of normal human pancreatic duct epithelial cells and the cancer cells. This review focuses on the regulation of tight junctions via protein kinase C during EMT in human pancreatic cancer for the purpose of developing new diagnostic and therapeutic modalities for pancreatic cancer.
Collapse
|
50
|
Patschan D, Müller GA. Acute kidney injury. J Inj Violence Res 2014; 7:19-26. [PMID: 25618438 PMCID: PMC4288292 DOI: 10.5249/jivr.v7i1.604] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 07/09/2014] [Indexed: 01/07/2023] Open
Abstract
Acute kidney injury is a frequent and serious complication in hospitalized patients. Mortality rates have not substantially been decreased during the last 20 years. In most patients AKI results from transient renal hypoperfusion or ischemia. The consequences include tubular cell dysfunction/damage, inflammation of the organ, and post-ischemic microvasculopathy. The two latter events perpetuate kidney damage in AKI. Clinical manifestations result from diminished excretion of water, electrolytes, and endogenous / exogenous waste products. Patients are endangered by cardiovascular complications such as hypertension, heart failure, and arrhythmia. In addition, the whole organism may be affected by systemic toxification (uremia). The diagnostic approach in AKI involves several steps with renal biopsy inevitable in some patients. The current therapy focuses on preventing further kidney damage and on treatment of complications. Different pharmacological strategies have failed to significantly improve prognosis in AKI. If dialysis treatment becomes mandatory, intermittent and continuous renal replacement therapies are equally effective. Thus, new therapies are urgently needed in order to reduce short- and long-term outcome in AKI. In this respect, stem cell-based regimens may offer promising perspectives.
Collapse
Affiliation(s)
| | - Gerhard Anton Müller
- Department of Nephrology and Rheumatology, University Medical Center of Göttingen, Göttingen, Germany. ,
| |
Collapse
|