51
|
Wang WJ, Cai GY, Chen XM. Cellular senescence, senescence-associated secretory phenotype, and chronic kidney disease. Oncotarget 2017; 8:64520-64533. [PMID: 28969091 PMCID: PMC5610023 DOI: 10.18632/oncotarget.17327] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022] Open
Abstract
Chronic kidney disease (CKD) is increasingly being accepted as a type of renal ageing. The kidney undergoes age-related alterations in both structure and function. To date, a comprehensive analysis of cellular senescence and senescence-associated secretory phenotype (SASP) in CKD is lacking. Hence, this review mainly discusses the relationship between the two phenomena to show the striking similarities between SASP and CKD-associated secretory phenotype (CASP). It has been reported that replicative senescence, stress-induced premature ageing, and epigenetic abnormalities participate in the occurrence and development of CKD. Genomic damage and external environmental stimuli cause increased levels of oxidative stress and a chronic inflammatory state as a result of irreversible cell cycle arrest and low doses of SASP. Similar to SASP, CASP factors activate tissue repair by multiple mechanisms. Once tissue repair fails, the accumulated SASP or CASP species aggravate DNA damage response (DDR) and cause the senescent cells to secrete more SASP factors, accelerating the process of cellular ageing and eventually leading to various ageing-related changes. It is concluded that cellular senescence and SASP participate in the pathological process of CKD, and correspondingly CKD accelerated the progression of cell senescence and the secretion of SASP. These results will facilitate the integration of these mechanisms into the care and management of CKD and other age-related diseases.
Collapse
Affiliation(s)
- Wen-Juan Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
- Department of Nephrology, Beijing Changping Hospital, Beijing 102200, China
| | - Guang-Yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| |
Collapse
|
52
|
Masum MA, Ichii O, Elewa YHA, Nakamura T, Kon Y. Local CD34-positive capillaries decrease in mouse models of kidney disease associating with the severity of glomerular and tubulointerstitial lesions. BMC Nephrol 2017; 18:280. [PMID: 28870174 PMCID: PMC5584339 DOI: 10.1186/s12882-017-0694-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/21/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The renal vasculature plays important roles in both homeostasis and pathology. In this study, we examined pathological changes in the renal microvascular in mouse models of kidney diseases. METHODS Glomerular lesions (GLs) in autoimmune disease-prone male BXSB/MpJ-Yaa (Yaa) mice and tubulointerstitial lesions (TILs) in male C57BL/6 mice subjected to unilateral ureteral obstruction (UUO) for 7 days were studied. Collected kidneys were examined using histopathological techniques. A nonparametric Mann-Whitney U test (P < 0.05) was performed to compare healthy controls and the experimental mice. The Kruskal-Wallis test was used to compare three or more groups, and multiple comparisons were performed using Scheffe's method when significant differences were observed (P < 0.05). RESULTS Yaa mice developed severe autoimmune glomerulonephritis, and the number of CD34+ glomerular capillaries decreased significantly in GLs compared to that in control mice. However, UUO-treated mice showed severe TILs only, and CD34+ tubulointerstitial capillaries were decreased significantly in TILs with the progression of tubulointerstitial fibrosis compared to those in untreated control kidneys. Infiltrations of B-cells, T-cells, and macrophages increased significantly in the respective lesions of both disease models (P < 0.05). In observations of vascular corrosion casts by scanning electron microscopy and of microfil rubber-perfused thick kidney sections by fluorescence microscopy, segmental absences of capillaries were observed in the GLs and TILs of Yaa and UUO-treated mice, respectively. Further, transmission electron microscopy revealed capillary endothelial injury in the respective lesions of both models. The numbers of CD34+ glomerular and tubulointerstitial capillaries were negatively correlated with all examined parameters in GLs (P < 0.05) and TILs (P < 0.01), respectively. CONCLUSIONS From the analysis of mouse models, we identified inverse pathological correlations between the number of local capillaries in GLs and TILs and the severity of kidney diseases.
Collapse
Affiliation(s)
- Md Abdul Masum
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
| | - Yaser Hosny Ali Elewa
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
- Department of Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Teppei Nakamura
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
- Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Tokyo, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
| |
Collapse
|
53
|
Zhu F, Chong Lee Shin OLS, Pei G, Hu Z, Yang J, Zhu H, Wang M, Mou J, Sun J, Wang Y, Yang Q, Zhao Z, Xu H, Gao H, Yao W, Luo X, Liao W, Xu G, Zeng R, Yao Y. Adipose-derived mesenchymal stem cells employed exosomes to attenuate AKI-CKD transition through tubular epithelial cell dependent Sox9 activation. Oncotarget 2017; 8:70707-70726. [PMID: 29050313 PMCID: PMC5642588 DOI: 10.18632/oncotarget.19979] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/18/2017] [Indexed: 12/19/2022] Open
Abstract
Acute kidney injury (AKI) predisposes patients to an increased risk into progressive chronic kidney disease (CKD), however effective treatments are still elusive. This study aimed to investigate the therapeutic efficacy of human adipose-derived MSCs (hAD-MSCs) in the prevention of AKI-CKD transition, and illuminate the role of Sox9, a vital transcription factor in the development of kidney, in this process. C57BL/6 mice were subjected to unilateral renal ischemia/reperfusion (I/R) with or without hAD-MSC treatment. We found that hAD-MSC treatment upregulated the expression of tubular Sox9, promoted tubular regeneration, attenuated AKI, and mitigated subsequent renal fibrosis. However, these beneficial effects were abolished by a drug inhibiting the release of exosomes from hAD-MSCs. Similarly, Sox9 inhibitors reversed these protective effects. Further, we verified that hAD-MSCs activated tubular Sox9 and prevented TGF-β1-induced transformation of TECs into pro-fibrotic phenotype through exosome shuttling in vitro, but the cells did not inhibit TGF-β1-induced transition of fibroblasts into myofibroblasts. Inhibiting the release of exosomes from hAD-MSCs or the expression of Sox9 in TECs reversed these antifibrotic effects. In conclusion, hAD-MSCs employed exosomes to mitigate AKI-CKD transition through tubular epithelial cell dependent activation of Sox9.
Collapse
Affiliation(s)
- Fengming Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Octavia L S Chong Lee Shin
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Guangchang Pei
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhizhi Hu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Juan Yang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Han Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Meng Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jingyi Mou
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jie Sun
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yuxi Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Qian Yang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhi Zhao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Huzi Xu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Hui Gao
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Weiqi Yao
- Wuhan Institute of Biotechnology, Guanggu Biolake, Wuhan 430000, Hubei, China
| | - Xiao Luo
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wenhui Liao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Gang Xu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Rui Zeng
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ying Yao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
54
|
Rysz J, Gluba-Brzózka A, Franczyk B, Jabłonowski Z, Ciałkowska-Rysz A. Novel Biomarkers in the Diagnosis of Chronic Kidney Disease and the Prediction of Its Outcome. Int J Mol Sci 2017; 18:E1702. [PMID: 28777303 PMCID: PMC5578092 DOI: 10.3390/ijms18081702] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/17/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
In its early stages, symptoms of chronic kidney disease (CKD) are usually not apparent. Significant reduction of the kidney function is the first obvious sign of disease. If diagnosed early (stages 1 to 3), the progression of CKD can be altered and complications reduced. In stages 4 and 5 extensive kidney damage is observed, which usually results in end-stage renal failure. Currently, the diagnosis of CKD is made usually on the levels of blood urea and serum creatinine (sCr), however, sCr has been shown to be lacking high predictive value. Due to the development of genomics, epigenetics, transcriptomics, proteomics, and metabolomics, the introduction of novel techniques will allow for the identification of novel biomarkers in renal diseases. This review presents some new possible biomarkers in the diagnosis of CKD and in the prediction of outcome, including asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA), uromodulin, kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), miRNA, ncRNA, and lincRNA biomarkers and proteomic and metabolomic biomarkers. Complicated pathomechanisms of CKD development and progression require not a single marker but their combination in order to mirror all types of alterations occurring in the course of this disease. It seems that in the not so distant future, conventional markers may be exchanged for new ones, however, confirmation of their efficacy, sensitivity and specificity as well as the reduction of analysis costs are required.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, WAM Teaching Hospital, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Zbigniew Jabłonowski
- I Department of Urology, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Aleksandra Ciałkowska-Rysz
- Palliative Medicine Unit, Chair of Oncology, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| |
Collapse
|
55
|
Heiss RU, Fahlbusch FB, Jacobi J, Daniel C, Ekici AB, Cordasic N, Amann K, Hartner A, Hilgers KF. Blunted transcriptional response to skeletal muscle ischemia in rats with chronic kidney disease: potential role for impaired ischemia-induced angiogenesis. Physiol Genomics 2017; 49:230-237. [PMID: 28213570 DOI: 10.1152/physiolgenomics.00124.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/15/2017] [Indexed: 11/22/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with increased cardiovascular morbidity and mortality. Previous studies indicated an impairment of ischemia-induced angiogenesis in skeletal muscle of rats with CKD. We performed a systematic comparison of early gene expression in response to ischemia in rats with or without CKD to identify potential molecular mechanisms underlying impaired angiogenesis in CKD. CKD was induced in male rats by 5/6 nephrectomy (SNX); control rats were sham operated (sham). Eight weeks later, ischemia of the right limb was induced by ligation and resection of the femoral artery. Rats were killed 24 h after the onset of ischemia, and RNA was extracted from the musculus soleus of the ischemic and the nonischemic hindlimb. To identify differentially expressed transcripts, we analyzed RNA with Affymetrix GeneChip Rat Genome 230 2.0 Arrays. RT-PCR analysis of selected genes was performed to validate observed changes. Hindlimb ischemia upregulated 239 genes in CKD and 299 genes in control rats (66% overlap), whereas only a few genes were downregulated (14 in CKD and 34 in controls) compared with the nonischemic limb of the same animals. Comparison between the ischemic limbs of CKD and controls revealed downregulation of 65 genes in CKD; 37 of these genes were also among the ischemia-induced genes in controls. Analysis of functional groups (other than angiogenesis) pointed to genes involved in leukocyte recruitment and fatty acid metabolism. Transcript expression profiling points to a relatively small number of differentially expressed genes that may underlie the impaired postischemic angiogenesis in CKD.
Collapse
Affiliation(s)
- Rafael U Heiss
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany.,Department of Radiology, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Fabian B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Johannes Jacobi
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Christoph Daniel
- Institute for Nephropathology, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany; and
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Nada Cordasic
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Kerstin Amann
- Institute for Nephropathology, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany; and
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Karl F Hilgers
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany;
| |
Collapse
|
56
|
Khurana R, Ranches G, Schafferer S, Lukasser M, Rudnicki M, Mayer G, Hüttenhofer A. Identification of urinary exosomal noncoding RNAs as novel biomarkers in chronic kidney disease. RNA (NEW YORK, N.Y.) 2017; 23:142-152. [PMID: 27872161 PMCID: PMC5238789 DOI: 10.1261/rna.058834.116] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/08/2016] [Indexed: 06/06/2023]
Abstract
In chronic kidney disease (CKD), the decline in the glomerular filtration rate is associated with increased morbidity and mortality and thus poses a major challenge for healthcare systems. While the contribution of tissue-derived miRNAs and mRNAs to CKD progression has been extensively studied, little is known about the role of urinary exosomes and their association with CKD. Exosomes are small, membrane-derived endocytic vesicles that contribute to cell-to-cell communication and are present in various body fluids, such as blood or urine. Next-generation sequencing approaches have revealed that exosomes are enriched in noncoding RNAs and thus exhibit great potential for sensitive nucleic acid biomarkers in various human diseases. Therefore, in this study we aimed to identify urinary exosomal ncRNAs as novel biomarkers for diagnosis of CKD. Since up to now most approaches have focused on the class of miRNAs, we extended our analysis to several other noncoding RNA classes, such as tRNAs, tRNA fragments (tRFs), mitochondrial tRNAs, or lincRNAs. For their computational identification from RNA-seq data, we developed a novel computational pipeline, designated as ncRNASeqScan. By these analyses, in CKD patients we identified 30 differentially expressed ncRNAs, derived from urinary exosomes, as suitable biomarkers for early diagnosis. Thereby, miRNA-181a appeared as the most robust and stable potential biomarker, being significantly decreased by about 200-fold in exosomes of CKD patients compared to healthy controls. Using a cell culture system for CKD indicated that urinary exosomes might indeed originate from renal proximal tubular epithelial cells.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers/urine
- Case-Control Studies
- Early Diagnosis
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Exosomes/chemistry
- Exosomes/metabolism
- Female
- Glomerular Filtration Rate
- High-Throughput Nucleotide Sequencing
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Male
- MicroRNAs/urine
- Middle Aged
- Molecular Sequence Annotation
- RNA/urine
- RNA, Long Noncoding/urine
- RNA, Mitochondrial
- RNA, Transfer/urine
- Renal Insufficiency, Chronic/diagnosis
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/urine
- Sequence Analysis, RNA
- Severity of Illness Index
Collapse
Affiliation(s)
- Rimpi Khurana
- Division of Genomics and RNomics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Glory Ranches
- Division of Genomics and RNomics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Simon Schafferer
- Division of Genomics and RNomics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Melanie Lukasser
- Division of Genomics and RNomics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Michael Rudnicki
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Gert Mayer
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Alexander Hüttenhofer
- Division of Genomics and RNomics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
- i-med GenomeSeq Core, 6020 Innsbruck, Austria
| |
Collapse
|
57
|
Liu M, Ning X, Li R, Yang Z, Yang X, Sun S, Qian Q. Signalling pathways involved in hypoxia-induced renal fibrosis. J Cell Mol Med 2017; 21:1248-1259. [PMID: 28097825 PMCID: PMC5487923 DOI: 10.1111/jcmm.13060] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 11/18/2016] [Indexed: 12/23/2022] Open
Abstract
Renal fibrosis is the common pathological hallmark of progressive chronic kidney disease (CKD) with diverse aetiologies. Recent researches have highlighted the critical role of hypoxia during the development of renal fibrosis as a final common pathway in end‐stage kidney disease (ESKD), which joints the scientist's attention recently to exploit the molecular mechanism underlying hypoxia‐induced renal fibrogenesis. The scaring formation is a multilayered cellular response and involves the regulation of multiple hypoxia‐inducible signalling pathways and complex interactive networks. Therefore, this review will focus on the signalling pathways involved in hypoxia‐induced pathogenesis of interstitial fibrosis, including pathways mediated by HIF, TGF‐β, Notch, PKC/ERK, PI3K/Akt, NF‐κB, Ang II/ROS and microRNAs. Roles of molecules such as IL‐6, IL‐18, KIM‐1 and ADO are also reviewed. A comprehensive understanding of the roles that these hypoxia‐responsive signalling pathways and molecules play in the context of renal fibrosis will provide a foundation towards revealing the underlying mechanisms of progression of CKD and identifying novel therapeutic targets. In the future, promising new effective therapy against hypoxic effects may be successfully translated into the clinic to alleviate renal fibrosis and inhibit the progression of CKD.
Collapse
Affiliation(s)
- Minna Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Rong Li
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhen Yang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaoxia Yang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qi Qian
- Department of Medicine, Division of Nephrology and hypertension, Mayo Clinic College of Medicine, Mayo Graduate School, Rochester, MN, USA
| |
Collapse
|
58
|
Burkhardt D, Bartosova M, Schaefer B, Grabe N, Lahrmann B, Nasser H, Freise C, Schneider A, Lingnau A, Degenhardt P, Ranchin B, Sallay P, Cerkauskiene R, Malina M, Ariceta G, Schmitt CP, Querfeld U. Reduced Microvascular Density in Omental Biopsies of Children with Chronic Kidney Disease. PLoS One 2016; 11:e0166050. [PMID: 27846250 PMCID: PMC5113061 DOI: 10.1371/journal.pone.0166050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/21/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Endothelial dysfunction is an early manifestation of cardiovascular disease (CVD) and consistently observed in patients with chronic kidney disease (CKD). We hypothesized that CKD is associated with systemic damage to the microcirculation, preceding macrovascular pathology. To assess the degree of "uremic microangiopathy", we have measured microvascular density in biopsies of the omentum of children with CKD. PATIENTS AND METHODS Omental tissue was collected from 32 healthy children (0-18 years) undergoing elective abdominal surgery and from 23 age-matched cases with stage 5 CKD at the time of catheter insertion for initiation of peritoneal dialysis. Biopsies were analyzed by independent observers using either a manual or an automated imaging system for the assessment of microvascular density. Quantitative immunohistochemistry was performed for markers of autophagy and apoptosis, and for the abundance of the angiogenesis-regulating proteins VEGF-A, VEGF-R2, Angpt1 and Angpt2. RESULTS Microvascular density was significantly reduced in uremic children compared to healthy controls, both by manual imaging with a digital microscope (median surface area 0.61% vs. 0.95%, p<0.0021 and by automated quantification (total microvascular surface area 0.89% vs. 1.17% p = 0.01). Density measured by manual imaging was significantly associated with age, height, weight and body surface area in CKD patients and healthy controls. In multivariate analysis, age and serum creatinine level were the only independent, significant predictors of microvascular density (r2 = 0.73). There was no immunohistochemical evidence for apoptosis or autophagy. Quantitative staining showed similar expression levels of the angiogenesis regulators VEGF-A, VEGF-receptor 2 and Angpt1 (p = 0.11), but Angpt2 was significantly lower in CKD children (p = 0.01). CONCLUSIONS Microvascular density is profoundly reduced in omental biopsies of children with stage 5 CKD and associated with diminished Angpt2 signaling. Microvascular rarefaction could be an early systemic manifestation of CKD-induced cardiovascular disease.
Collapse
Affiliation(s)
- Dorothea Burkhardt
- Department of Pediatric Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Niels Grabe
- Bioquant, Hamamatsu Tissue Imaging and Analysis (TIGA) Center, University of Heidelberg, Heidelberg, Germany
| | - Bernd Lahrmann
- Bioquant, Hamamatsu Tissue Imaging and Analysis (TIGA) Center, University of Heidelberg, Heidelberg, Germany
| | - Hamoud Nasser
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Freise
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Schneider
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Lingnau
- Department of Pediatric Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Degenhardt
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Surgery, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Bruno Ranchin
- Hospices Civils de Lyon, Service de Nephrologie Pediatrique and Epicime-Centre d’Investigation Clinique 1407, Hopital Femme Mere Enfant, Lyon, France
| | - Peter Sallay
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Rimante Cerkauskiene
- Coordinating Centre for Children’s Rare Diseases, Children´s Hospital, Affiliate of Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Michal Malina
- Department of Pediatrics, Second Faculty of Medicine, Charles University-Prague, Prague 5, Czech Republic
| | - Gema Ariceta
- Servicio de Nefrología Pediátrica, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Uwe Querfeld
- Department of Pediatric Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
59
|
Sezer M, Kocaaga M, Aslanger E, Atici A, Demirkiran A, Bugra Z, Umman S, Umman B. Bimodal Pattern of Coronary Microvascular Involvement in Diabetes Mellitus. J Am Heart Assoc 2016; 5:JAHA.116.003995. [PMID: 27930353 PMCID: PMC5210326 DOI: 10.1161/jaha.116.003995] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background The contribution of functionally disturbed coronary autoregulation and structurally impaired microvascular vasodilatory function to reduced coronary flow velocity reserve, reflecting impaired coronary microcirculation in diabetes mellitus (DM), has not been clearly elucidated. The objective of this study was to identify the mechanism of coronary microvascular impairment in DM in relation to duration of disease. Methods and Results Coronary flow velocities in the anterior descending coronary artery were assessed by transthoracic echocardiography following angiography revealing normal epicardial coronary arteries in 55 diabetic and 47 nondiabetic patients. Average peak flow velocities, coronary flow velocity reserve, and microvascular resistance in baseline and hyperemic conditions (baseline and hyperemic microvascular resistance, respectively) were assessed. Reduced coronary flow velocity reserve in patients with short duration (<10 years) of DM compared with nondiabetic patients was primarily driven by increased baseline average peak flow velocity (26.50±5.6 versus 22.08±4.31, P=0.008) in the presence of decreased baseline microvascular resistance (3.69±0.86 versus 4.34±0.76, P=0.003). In contrast, decreased coronary flow velocity reserve in patients with long‐standing (≥10 years) DM compared with nondiabetic patients was predominantly driven by reduced hyperemic average peak flow velocity (41.57±10.01 versus 53.47±11.8, P<0.001) due to increased hyperemic microvascular resistance (2.13±0.42 versus 1.69±0.39, P<0.001). Conclusions Both altered coronary autoregulation and impaired microvascular vasodilatory function contribute to DM‐related coronary microvascular impairment in a time‐dependent manner. DM‐induced early functional microvascular autoregulatory impairment seems to evolve into structural microvascular impairment in the initially overperfused microvascular territory at the later stage of disease.
Collapse
Affiliation(s)
- Murat Sezer
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mehmet Kocaaga
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Emre Aslanger
- Department of Cardiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Adem Atici
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ahmet Demirkiran
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Zehra Bugra
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sabahattin Umman
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Berrin Umman
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
60
|
Relation of peritubular capillary features to class of lupus nephritis. BMC Nephrol 2016; 17:169. [PMID: 27829387 PMCID: PMC5103390 DOI: 10.1186/s12882-016-0388-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/03/2016] [Indexed: 02/05/2023] Open
Abstract
Background Experimental studies have linked peritubular capillary (PTC) loss with progression of chronic kidney disease. Minimal information on PTC in lupus nephritis (LN) has been reported. We therefore evaluated the PTC area in different classes of LN and determined if specific clinical characteristics correlated with PTC changes. Methods Renal biopsies of 253 subjects with LN (categorized using the ISN/RPS 2003 classification) and 13 normal renal donors (the controls) were retrospectively evaluated for PTC morphology by staining for CD31 with immunohistochemistry method. The percent positive area of PTC (% PTC) was correlated with serum and urinary measures of renal function and renal pathology. Results Significant PTC loss was observed in all classes of LN compared to controls. The % PTC area was highest in controls (7.64±1.48 %) with levels of 1.95±1.50, 4.16±3.85, 4.19±4.45, 5.02±1.79, and 4.45±3.75 in classes II, III, IV, IV combined with V and V, respectively (all p values < 0.05). The lowest PTC density was observed in class II LN, but this may be because some cases with worse classes of LN showed increased PTC density due to abnormally dilated capillaries associated with acute inflammation and angiogenesis. %PTC was increased in those with hematuria (5.8±5.2 vs. 3.6±3.4 %, red blood cells 3-10 vs. < 3 cells/high power field, p < 0.05) and was reduced in those with a moderately declined renal function (3.29±3.40 vs. 4.42±4.12, eGFR 15-59 vs. ≥ 60 ml/min/1.73 m2, p < 0.05). Nephrotic-range proteinuria also trended to be associated with lower PTC density although it did not reach statistical significance (3.1±2.6 vs. 4.9±4.5, p= 0.067). Conclusions LN is associated with PTC loss and the severity correlates with reduced renal function. Further studies are needed to investigate whether a loss of PTC can predict long term renal outcomes in LN. Electronic supplementary material The online version of this article (doi:10.1186/s12882-016-0388-2) contains supplementary material, which is available to authorized users.
Collapse
|
61
|
Gewin L, Zent R, Pozzi A. Progression of chronic kidney disease: too much cellular talk causes damage. Kidney Int 2016; 91:552-560. [PMID: 27773427 DOI: 10.1016/j.kint.2016.08.025] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/31/2016] [Accepted: 08/16/2016] [Indexed: 01/10/2023]
Abstract
Tubulointerstitial fibrosis, tubular atrophy, and peritubular capillary rarefaction are major hallmarks of chronic kidney disease. The tubulointerstitium consists of multiple cell components including tubular epithelial, mesenchymal (fibroblasts and pericytes), endothelial, and inflammatory cells. Crosstalk among these cell components is a key component in the pathogenesis of this complex disease. After severe or recurrent injury, the renal tubular epithelial cells undergo changes in structure and cell cycle that are accompanied by altered expression and production of cytokines. These cytokines contribute to the initiation of the fibrotic response by favoring activation of fibroblasts, recruitment of inflammatory cells, and loss of endothelial cells. This review focuses on how augmented growth factor and cytokine production induces epithelial crosstalk with cells in the interstitium to promote progressive tubulointerstitial fibrosis after renal injury.
Collapse
Affiliation(s)
- Leslie Gewin
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA.
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Molecular Physiology and Biophysics, Vanderbilt Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
62
|
Zou X, Gu D, Xing X, Cheng Z, Gong D, Zhang G, Zhu Y. Human mesenchymal stromal cell-derived extracellular vesicles alleviate renal ischemic reperfusion injury and enhance angiogenesis in rats. Am J Transl Res 2016; 8:4289-4299. [PMID: 27830012 PMCID: PMC5095321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/14/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) derived extracellular vesicles (EVs) were regarded as a potent medium for kidney injury repair and angiogenesis were regarded as an important step in tissue regeneration. However, the pro-angiogenesis effect of MSC-EVs in ischemia-reperfusion induced kidney injury and its potential mechanisms have yet to be determined. METHODS EVs were isolated from the medium of human umbilical cord-derived MSCs (huMSCs) were injected in rats intravenously after unilateral kidney ischemia. Animals were sacrificed at 24 h and 2 weeks after injury. The renal functions and histology staining were examined to assess the therapeutic effect of the EVs. Moreover, we investigated the pro-angiogenesis effects of EVs in injured kidneys and tested the angiogenesis-related factors to further illuminate the probable mechanisms. RESULTS It was observed that EVs could reduce cell apoptosis and enhances proliferation 24 h after kidney injury, meanwhile renal function was improved and the histological lesion was mitigated. Moreover, renal VEGF was up-regulated by EVs and HIF-1α was down-regulated. Further, the increase of capillary vessel density and reduce of renal fibrosis was observed after 2 weeks. In vitro, EVs could deliver human VEGF directly to renal tubular epithelial cells (TECs) and increase VEGF levels. Most important, all the beneficial effects of EVs were abrogated by RNase treated except for the delivery of human VEGF. CONCLUSIONS Human MSC-EVs could protect against ischemic/reperfusion injury induced kidney injury through pro-angiogenesis effects in HIF-1α independent manner, and both the delivery of pro-angiogenesis related VEGF and RNAs were involved in this process.
Collapse
Affiliation(s)
- Xiangyu Zou
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
- Department of Urology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Di Gu
- Department of Urology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Xiaoyu Xing
- Department of Urology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | | | - Dongliang Gong
- Department of Urology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Guangyuan Zhang
- Department of Urology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
- Department of Urology, Affiliated Zhongda Hospital of Southeast UniversityNanjing, China
| | - Yingjian Zhu
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
63
|
Daly KP, Stack M, Eisenga MF, Keane JF, Zurakowski D, Blume ED, Briscoe DM. Vascular endothelial growth factor A is associated with the subsequent development of moderate or severe cardiac allograft vasculopathy in pediatric heart transplant recipients. J Heart Lung Transplant 2016; 36:434-442. [PMID: 27865734 DOI: 10.1016/j.healun.2016.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/08/2016] [Accepted: 09/16/2016] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) is the leading cause of chronic allograft loss after pediatric heart transplantation. We hypothesized that biomarkers of endothelial injury and repair would predict CAV development in pediatric heart transplant recipients. METHODS Blood was collected from pediatric heart transplant recipients at the time of routine annual coronary angiography, and the concentrations of 13 angiogenesis-related molecules were determined. The primary end point was the presence of moderate or severe CAV by angiography during a 5-year follow-up period. RESULTS The study enrolled 48 recipients (57% male) with a median age of 15.5 years (range, 2-22 years) and median time post-transplant of 5.8 years (range, 2-15 years). Eight recipients developed moderate/severe CAV at a median follow-up of 4.7 years, of whom 3 died, 3 underwent retransplantation, 1 had a myocardial infarction, and 1 was listed for retransplantation. Clinical characteristics associated with the development of moderate/severe CAV included prednisone use at enrollment (p = 0.03) and positive recipient cytomegalovirus immunoglobulin G at the time of transplant (p = < 0.01). Multivariable Cox proportional hazards regression identified plasma vascular endothelial growth factor (VEGF)-A concentration greater than 90 pg/ml at the time of blood draw as a significant predictor of time to moderate or severe CAV (hazard ratio, 14.3; 95% confidence interval, 1.3-163). Receiver operating characteristic curve analysis demonstrated that VEGF-A shows moderate performance for association with the subsequent development of CAV (area under the curve, 0.77; 95% confidence interval, 0.61-0.92). CONCLUSIONS VEGF-A levels in pediatric heart transplant recipients are associated with clinically important CAV progression within the subsequent 5 years.
Collapse
Affiliation(s)
- Kevin P Daly
- Transplant Research Program, Department of Medicine, Boston, Massachusetts; Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Maria Stack
- Transplant Research Program, Department of Medicine, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Nephrology, Department of Medicine, Boston, Massachusetts
| | - Michele F Eisenga
- Transplant Research Program, Department of Medicine, Boston, Massachusetts; Division of Nephrology, Department of Medicine, Boston, Massachusetts
| | - John F Keane
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - David Zurakowski
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Department of Anesthesia, Boston Children's Hospital, Boston, Massachusetts; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth D Blume
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - David M Briscoe
- Transplant Research Program, Department of Medicine, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Nephrology, Department of Medicine, Boston, Massachusetts.
| |
Collapse
|
64
|
Hamar P, Kerjaschki D. Blood capillary rarefaction and lymphatic capillary neoangiogenesis are key contributors to renal allograft fibrosis in an ACE inhibition rat model. Am J Physiol Heart Circ Physiol 2016; 311:H981-H990. [PMID: 27496878 DOI: 10.1152/ajpheart.00320.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/29/2016] [Indexed: 12/17/2022]
Abstract
Chronic allograft fibrosis is the major cause of graft loss in kidney transplantation. Progression can only be reduced by inhibition of the renin-angiotensin system (RAS). We tested the hypothesis that the protection provided by angiotensin-converting enzyme (ACE) inhibition also decreases capillary rarefaction, lymphangiogenesis, and podocyte injury in allograft fibrosis. Fisher kidneys were transplanted into bilaterally nephrectomized Lewis rats treated with enalapril (60 mg/kg per day) (ACE inhibitor, ACEi) or vehicle. Proteinuria, blood urea nitrogen, and plasma creatinine were regularly assessed, and grafts were harvested for morphological and immunohistological analysis at various times up to 32 wk. In the vehicle group, many new lymphatic capillaries and severe and diffuse mononuclear infiltration of allografts were observed already 1 wk after transplantation. Lymphangiogenesis increased until week 4, by which time inflammatory infiltration became focal. Lymphatic capillaries were often located at sites of inflammation. Progressive interstitial fibrosis, glomerulosclerosis, capillary rarefaction, and proteinuria appeared later, at weeks 4-12 The number of lymphatic capillary cross sections strongly correlated with the interstitial fibrosis score. Podoplanin immunostaining, a marker of healthy podocytes, disappeared from inflamed or sclerotic glomerular areas. ACEi protected from lymphangiogenesis and associated inflammation, preserved glomerular podoplanin protein expression, and reduced glomerulosclerosis, proteinuria, tubulointerstitial fibrosis, and blood capillary rarefaction at 32 wk. In conclusion, ACEi considerably decreased and/or delayed both glomerulosclerosis and tubulointerstitial injury. Prevention of glomerular podoplanin loss and proteinuria could be attributed to the known intraglomerular pressure-lowering effects of ACEi. Reduction of lymphangiogenesis could contribute to amelioration of tubulointerstitial fibrosis and inflammatory infiltration after ACEi.
Collapse
Affiliation(s)
- Péter Hamar
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary; and
| | | |
Collapse
|
65
|
Jia X, Pan J, Li X, Li N, Han Y, Feng X, Cui J. Bone marrow mesenchymal stromal cells ameliorate angiogenesis and renal damage via promoting PI3k-Akt signaling pathway activation in vivo. Cytotherapy 2016; 18:838-45. [PMID: 27210720 DOI: 10.1016/j.jcyt.2016.03.300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/11/2016] [Accepted: 03/31/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The objective of this study was to investigate the effects of the intravenous transplantation of bone marrow mesenchymal stromal cells (BM-MSCs) on the repair of glomerular endothelia and angiogenesis in rats with chronic renal failure (CRF). Furthermore, the mechanism of BM-MSCs promoting angiogenesis was explored by detection of Akt and P-Akt protein expression in rat kidney tissue. MATERIAL AND METHODS A rat model with CRF was established by adenine. Immature male Wistar rats were randomly divided into control group, model group and treatment group. Model group rats were injected with phosphate-buffered saline (PBS) via tail vein 24 h after the successful modeling, whereas the treatment group rats were injected with BM-MSCs. Eight weeks later, urine and blood were collected to assess 24-h proteinuria, serum creatinine (Scr) and blood urea nitrogen (BUN). We identified glomerular capillaries density using JG12 immunostaining. Levels of vascular endothelial growth factor (VEGF) were assayed using enzyme-linked immunosorbant assay (ELISA). We used Western blot to determine protein expression of p-Akt and Akt in renal tissues. RESULTS Adenine induced chronic renal damage, as indicated by the mass proteinuria, deterioration of renal function and the histopathologic injury in tubules and interstitium. BM-MSCs signficantly increased capillary density and improved renal function and serum VEGF. Additionally, activation of Akt (i.e., P-Akt significantly increased) in the treatment group was increased obviously. CONCLUSION BM-MSCs could alleviate the renal damages of adenine-induced CRF, reduce the excretion of proteinuria, increase the glomerular capillaries density, promote the secretion of VEGF and finally contribute to improve renal function. VEGF-induced angiogenesis is mediated through activating PI3k-Akt signaling pathway.
Collapse
Affiliation(s)
- Xiaojing Jia
- Department of Pediatric, Shanxi Medical University, Taiyuan, China
| | - Jinbing Pan
- Department of Pathology, Shanxi Provincial Children's Hospital, Taiyuan, China
| | - Xiuhua Li
- Department of Public Health, Shanxi Medical University, Taiyuan, China
| | - Na Li
- Department of Pediatric, Shanxi Medical University, Taiyuan, China
| | - Yan Han
- Department of Pediatric, Shanxi Medical University, Taiyuan, China
| | - Xing Feng
- Department of Pediatric, Shanxi Medical University, Taiyuan, China
| | - Jianjun Cui
- Department of Nephrology, Shanxi Provincial Children's Hospital, NO. 15, Xinmin North Street, Xinghualing District, Taiyuan City, Shanxi Province, China.
| |
Collapse
|
66
|
Abstract
The longstanding focus in chronic kidney disease (CKD) research has been on the glomerulus, which is sensible because this is where glomerular filtration occurs, and a large proportion of progressive CKD is associated with significant glomerular pathology. However, it has been known for decades that tubular atrophy is also a hallmark of CKD and that it is superior to glomerular pathology as a predictor of glomerular filtration rate decline in CKD. Nevertheless, there are vastly fewer studies that investigate the causes of tubular atrophy, and fewer still that identify potential therapeutic targets. The purpose of this review is to discuss plausible mechanisms of tubular atrophy, including tubular epithelial cell apoptosis, cell senescence, peritubular capillary rarefaction and downstream tubule ischemia, oxidative stress, atubular glomeruli, epithelial-to-mesenchymal transition, interstitial inflammation, lipotoxicity and Na(+)/H(+) exchanger-1 inactivation. Once a a better understanding of tubular atrophy (and interstitial fibrosis) pathophysiology has been obtained, it might then be possible to consider tandem glomerular and tubular therapeutic strategies, in a manner similar to cancer chemotherapy regimens, which employ multiple drugs to simultaneously target different mechanistic pathways.
Collapse
|
67
|
Park S, Kim JW, Kim JH, Lim CW, Kim B. Differential Roles of Angiogenesis in the Induction of Fibrogenesis and the Resolution of Fibrosis in Liver. Biol Pharm Bull 2016; 38:980-5. [PMID: 26133707 DOI: 10.1248/bpb.b15-00325] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Liver fibrosis is a wound healing process that includes inflammation, deposition of extracellular matrix molecules, and pathological neovascularization. Angiogenesis, which is defined by the formation of new blood vessels from pre-existing vessels, is a complex and dynamic process under both physiological and pathological conditions. Although whether angiogenesis can induce or occur in parallel with the progression of hepatic fibrosis has not yet been determined, intrahepatic sinusoidal formation and remodeling are key features of liver fibrosis. Some recent evidence has suggested that experimental inhibition of angiogenesis ameliorates the development of liver fibrosis, while other recent studies indicate that neutralization or genetic ablation of vascular endothelial growth factor (VEGF) in myeloid cells can delay tissue repair and fibrosis resolution in damaged liver. In this review, we briefly summarize the current knowledge about the differential roles of angiogenesis in the induction of fibrogenesis and the resolution of fibrosis in damaged livers. Possible strategies for the prevention and treatment of liver fibrosis are also discussed.
Collapse
Affiliation(s)
- Surim Park
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine
(BK21 Plus Program), Chonbuk National University
| | | | | | | | | |
Collapse
|
68
|
Saad A, Herrmann SM, Textor SC. Chronic renal ischemia in humans: can cell therapy repair the kidney in occlusive renovascular disease? Physiology (Bethesda) 2016; 30:175-82. [PMID: 25933818 DOI: 10.1152/physiol.00065.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Occlusive renovascular disease caused by atherosclerotic renal artery stenosis (ARAS) elicits complex biological responses that eventually lead to loss of kidney function. Recent studies indicate a complex interplay of oxidative stress, endothelial dysfunction, and activation of fibrogenic and inflammatory cytokines as a result of atherosclerosis, hypoxia, and renal hypoperfusion in this disorder. Human studies emphasize the limits of the kidney adaptation to reduced blood flow, eventually leading to renal hypoxia with activation of inflammatory and fibrogenic pathways. Several randomized prospective clinical trials show that stent revascularization alone in patients with atherosclerotic renal artery stenosis provides little additional benefit to medical therapy once these processes have developed and solidified. Experimental data now support developing adjunctive cell-based measures to support angiogenesis and anti-inflammatory renal repair mechanisms. These data encourage the study of endothelial progenitor cells and/or mesenchymal stem/stromal cells for the repair of damaged kidney tissue.
Collapse
Affiliation(s)
- Ahmed Saad
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
69
|
Rudnicki M, Beckers A, Neuwirt H, Vandesompele J. RNA expression signatures and posttranscriptional regulation in diabetic nephropathy. Nephrol Dial Transplant 2016. [PMID: 26209736 DOI: 10.1093/ndt/gfv079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In the last decade, the integration of molecular approaches including transcriptome and miRNome analyses uncovered pathological mechanisms involved in the progression of diabetic nephropathy (DN). Using these techniques, molecular marker candidates [both messenger RNA (mRNA) and miRNA] have also been identified which may enable the characterization of patients at high risk for progression to end-stage renal disease. The results of such studies are urgently needed for a molecular definition of DN and for targeted treatment to improve patient care. The heterogeneity of kidney tissue and the minute amounts of RNA isolated from renal biopsies remain a challenge for omics-studies. Nevertheless, several studies have succeeded in the identification of RNA expression signatures in patients with diabetes and kidney disease. These studies show a reduced expression of growth factors such as VEGF and EGF, and an increased expression of matrix components and matrix-modulating enzymes, an activation of specific NF-κB modules, inflammatory pathways and the complement system. microRNAs are involved in the fine-tuning of mRNA abundance by binding to the 3' untranslated region of a target mRNA, which leads in most cases to translational repression or mRNA cleavage and a decrease in protein output. Here, we review the platforms used for miRNA expression profiling and ways to predict miRNA targets and functions. Several miRNAs have been shown to be involved in the pathogenesis of DN (e.g. miR-21, miR-192, miR-215, miR-216a, miR-29, let-7, miR-25, miR-93, etc.). Functional studies provide evidence that miRNAs are not only diagnostic tools but also represent potential therapeutic targets in DN.
Collapse
Affiliation(s)
- Michael Rudnicki
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | | | - Hannes Neuwirt
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
70
|
Fu Q, Colgan SP, Shelley CS. Hypoxia: The Force that Drives Chronic Kidney Disease. Clin Med Res 2016; 14:15-39. [PMID: 26847481 PMCID: PMC4851450 DOI: 10.3121/cmr.2015.1282] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 09/30/2015] [Indexed: 12/15/2022]
Abstract
In the United States the prevalence of end-stage renal disease (ESRD) reached epidemic proportions in 2012 with over 600,000 patients being treated. The rates of ESRD among the elderly are disproportionally high. Consequently, as life expectancy increases and the baby-boom generation reaches retirement age, the already heavy burden imposed by ESRD on the US health care system is set to increase dramatically. ESRD represents the terminal stage of chronic kidney disease (CKD). A large body of evidence indicating that CKD is driven by renal tissue hypoxia has led to the development of therapeutic strategies that increase kidney oxygenation and the contention that chronic hypoxia is the final common pathway to end-stage renal failure. Numerous studies have demonstrated that one of the most potent means by which hypoxic conditions within the kidney produce CKD is by inducing a sustained inflammatory attack by infiltrating leukocytes. Indispensable to this attack is the acquisition by leukocytes of an adhesive phenotype. It was thought that this process resulted exclusively from leukocytes responding to cytokines released from ischemic renal endothelium. However, recently it has been demonstrated that leukocytes also become activated independent of the hypoxic response of endothelial cells. It was found that this endothelium-independent mechanism involves leukocytes directly sensing hypoxia and responding by transcriptional induction of the genes that encode the β2-integrin family of adhesion molecules. This induction likely maintains the long-term inflammation by which hypoxia drives the pathogenesis of CKD. Consequently, targeting these transcriptional mechanisms would appear to represent a promising new therapeutic strategy.
Collapse
Affiliation(s)
- Qiangwei Fu
- Kabara Cancer Research Institute, La Crosse, WI
| | - Sean P Colgan
- Mucosal Inflammation Program and University of Colorado School of Medicine, Aurora, CO
| | - Carl Simon Shelley
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
71
|
Rudnicki M, Perco P, D Haene B, Leierer J, Heinzel A, Mühlberger I, Schweibert N, Sunzenauer J, Regele H, Kronbichler A, Mestdagh P, Vandesompele J, Mayer B, Mayer G. Renal microRNA- and RNA-profiles in progressive chronic kidney disease. Eur J Clin Invest 2016; 46:213-26. [PMID: 26707063 DOI: 10.1111/eci.12585] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 12/20/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) contribute to chronic kidney disease (CKD) progression via regulating mRNAs involved in renal homeostasis. However, their association with clinical outcome remains poorly understood. MATERIALS AND METHODS We performed miRNA and mRNA expression profiling on renal biopsy sections by qPCR (miRNA) and microarrays (mRNA) in a discovery (n = 43) and in a validation (n = 29) cohort. miRNAs differentiating stable and progressive cases were inversely correlated with putative target mRNAs, which were further characterized by pathway analysis using KEGG pathways. RESULTS miR-30d, miR-140-3p, miR-532-3p, miR-194, miR-190, miR-204 and miR-206 were downregulated in progressive cases. These seven miRNAs correlated with upregulated 29 target mRNAs involved in inflammatory response, cell-cell interaction, apoptosis and intra-cellular signalling. In particular, miR-206 and miR-532-3p were associated with distinct biological processes via the expression of their target mRNAs: Reduced expression of miR-206 in progressive disease correlated with the upregulation of target mRNAs participating in inflammatory pathways (CCL19, CXCL1, IFNAR2, NCK2, PTK2B, PTPRC, RASGRP1 and TNFRSF25). Progressive cases also showed a lower expression of miR-532-3p and an increased expression of target transcripts involved in apoptosis pathways (MAP3K14, TNFRSF10B/TRAIL-R2, TRADD and TRAF2). In the validation cohort, we confirmed the decreased expression of miR-206 and miR-532-3p, and the inverse correlation of these miRNAs with the expression of nine of the 12 target genes. The levels of the identified miRNAs and the target mRNAs correlated with clinical parameters and histological damage indices. CONCLUSIONS These results suggest the involvement of specific miRNAs and mRNAs in biological pathways associated with the progression of CKD.
Collapse
Affiliation(s)
- Michael Rudnicki
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | - Paul Perco
- Emergentec Biodevelopment GmbH, Vienna, Austria
| | | | - Johannes Leierer
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Ninella Schweibert
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | - Judith Sunzenauer
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria.,Department of Nephrology, KH Elisabethinen, Linz, Austria
| | - Heinz Regele
- Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Andreas Kronbichler
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Bernd Mayer
- Emergentec Biodevelopment GmbH, Vienna, Austria
| | - Gert Mayer
- Department of Internal Medicine IV - Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
72
|
Olfert IM. Physiological Capillary Regression is not Dependent on Reducing VEGF Expression. Microcirculation 2016; 23:146-56. [PMID: 26660949 PMCID: PMC4744091 DOI: 10.1111/micc.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/03/2015] [Indexed: 01/04/2023]
Abstract
Investigations into physiologically controlled capillary regression report the provocative finding that microvessel regression occurs in the face of persistent elevation of skeletal muscle VEGF expression. TSP-1, a negative angiogenic regulator, is increasingly being observed to temporally correlate with capillary regression, suggesting that increased TSP-1 (and not reduction in VEGF per se) is needed to initiate, and likely regulate, capillary regression. Based on evidence being gleaned from physiologically mediated regression of capillaries, it needs to be recognized that capillary regression (and perhaps capillary rarefaction with disease) is not simply the reversal of factors used to stimulate angiogenesis. Rather, the conceptual understanding that angiogenesis and capillary regression each have specific and unique requirements that are biologically constrained to opposite sides of the balance between positive and negative angioregulatory factors may shed light on why anti-VEGF therapies have not lived up to the promise in reversing angiogenesis and providing the cure that many had hoped toward fighting cancer. Emerging evidence from physiological controlled angiogenesis suggest that cases involving excessive or uncontrolled capillary expansion may be best treated by therapies designed to increase expression of negative angiogenic regulators, whereas those involving capillary rarefaction may benefit from inhibiting negative regulators (like TSP-1).
Collapse
Affiliation(s)
- I Mark Olfert
- Division of Exercise Physiology, Center for Cardiovascular and Respiratory Sciences, Mary Babb Randolph Cancer Center, West Virginia Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
73
|
Stefanska A, Eng D, Kaverina N, Pippin JW, Gross KW, Duffield JS, Shankland SJ. Cells of renin lineage express hypoxia inducible factor 2α following experimental ureteral obstruction. BMC Nephrol 2016; 17:5. [PMID: 26746687 PMCID: PMC4706659 DOI: 10.1186/s12882-015-0216-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/22/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent studies indicate that mural cells of the preglomerular vessels, known as cells of renin lineage (CoRL), contribute to repair and regeneration of injured kidney glomeruli. However, their potential roles in tubulointerstitial disease are less understood. The aim of this study was to better understand CoRL number and distribution following UUO so that future mechanistic studies could be undertaken. METHODS We mapped the fate of CoRL in adult Ren1cCreER x Rs-tdTomato-R reporter mice that underwent UUO. Kidney biopsies from sham and UUO-subjected mice on days 3, 7, and 14 were evaluated by immunohistochemistry. RESULTS In sham animals, CoRL were restricted to juxtaglomerular location. At day 7 following UUO, CoRL increased two-fold, were perivascular in location, and co-expressed pericyte markers (PDGFßR, NG2), but did not express renin. At day 14 post UUO, labeled CoRL detached from vessels and were present in the interstitium, in areas of fibrosis, where they now expressed the myofibroblast marker alpha-smooth muscle actin. The increase in CoRL was likely due to proliferation as marked by BrdU labeling, and migration from the cortex. Following UUO starting from day 3, active hypoxia inducible factor-2α was detected in nuclei in labeled CoRL, in the cortex, but not those cells found in medulla. CONCLUSIONS We have demonstrated that arteriolar CoRL are potential kidney progenitors that may contribute to the initial vascular regeneration. However, in chronic kidney injury (≥14 days post UUO), perivascular CoRL transition to myofibroblast-like cells.
Collapse
Affiliation(s)
- Ania Stefanska
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, 98104, USA.
| | - Diana Eng
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, 98104, USA.
| | - Natalya Kaverina
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, 98104, USA.
| | - Jeffrey W Pippin
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, 98104, USA.
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| | | | - Stuart J Shankland
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, 98104, USA.
| |
Collapse
|
74
|
Pianta TJ, Peake PW, Pickering JW, Kelleher M, Buckley NA, Endre ZH. Evaluation of biomarkers of cell cycle arrest and inflammation in prediction of dialysis or recovery after kidney transplantation. Transpl Int 2015; 28:1392-404. [DOI: 10.1111/tri.12636] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/30/2014] [Accepted: 07/07/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Timothy J. Pianta
- Prince of Wales Clinical School; University of New South Wales; Sydney NSW Australia
- Northern Clinical School; Melbourne Medical School; University of Melbourne; Epping Vic Australia
| | - Philip W. Peake
- Prince of Wales Clinical School; University of New South Wales; Sydney NSW Australia
| | - John W. Pickering
- Department of Medicine; University of Otago; Christchurch New Zealand
| | - Michaela Kelleher
- Department of Nephrology; Prince of Wales Hospital; Sydney NSW Australia
| | | | - Zoltan H. Endre
- Prince of Wales Clinical School; University of New South Wales; Sydney NSW Australia
- Department of Medicine; University of Otago; Christchurch New Zealand
| |
Collapse
|
75
|
Silva JN, Polesskaya O, Wei HS, Rasheed IYD, Chamberlain JM, Nishimura C, Feng C, Dewhurst S. Chronic central nervous system expression of HIV-1 Tat leads to accelerated rarefaction of neocortical capillaries and loss of red blood cell velocity heterogeneity. Microcirculation 2015; 21:664-76. [PMID: 24813724 DOI: 10.1111/micc.12145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/06/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVES HIV-1 infection of the CNS is associated with impairment of CBF and neurocognitive function, and accelerated signs of aging. As normal aging is associated with rarefaction of the cerebral vasculature, we set out to examine chronic viral effects on the cerebral vasculature. METHODS DOX-inducible HIV-1 Tat-tg and WT control mice were used. Animals were treated with DOX for three weeks or five to seven months. Cerebral vessel density and capillary segment length were determined from quantitative image analyses of sectioned cortical tissue. In addition, movement of red blood cells in individual capillaries was imaged in vivo using multiphoton microscopy, to determine RBCV and flux. RESULTS Mean RBCV was not different between Tat-tg mice and age-matched WT controls. However, cortical capillaries from Tat-tg mice showed a significant loss of RBCV heterogeneity and increased RBCF that was attributed to a marked decrease in total cortical capillary length (35-40%) compared to WT mice. CONCLUSIONS Cerebrovascular rarefaction is accelerated in HIV-1 Tat-transgenic mice, and this is associated with alterations in red cell blood velocity. These changes may have relevance to the pathogenesis of HIV-associated neurocognitive disorders in an aging HIV-positive population.
Collapse
Affiliation(s)
- Jharon N Silva
- Departments of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Lavoz C, Alique M, Rodrigues-Diez R, Pato J, Keri G, Mezzano S, Egido J, Ruiz-Ortega M. Gremlin regulates renal inflammation via the vascular endothelial growth factor receptor 2 pathway. J Pathol 2015; 236:407-20. [PMID: 25810250 DOI: 10.1002/path.4537] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/03/2023]
Abstract
Inflammation is a main feature of progressive kidney disease. Gremlin binds to bone morphogenetic proteins (BMPs), acting as an antagonist and regulating nephrogenesis and fibrosis among other processes. Gremlin also binds to vascular endothelial growth factor receptor-2 (VEGFR2) in endothelial cells to induce angiogenesis. In renal cells, gremlin regulates proliferation and fibrosis, but there are no data about inflammatory-related events. We have investigated the direct effects of gremlin in the kidney, evaluating whether VEGFR2 is a functional gremlin receptor. Administration of recombinant gremlin to murine kidneys induced rapid and sustained activation of VEGFR2 signalling, located in proximal tubular epithelial cells. Gremlin bound to VEGFR2 in these cells in vitro, activating this signalling pathway independently of its action as an antagonist of BMPs. In vivo, gremlin caused early renal damage, characterized by activation of the nuclear factor (NF)-κB pathway linked to up-regulation of pro-inflammatory factors and infiltration of immune inflammatory cells. VEGFR2 blockade diminished gremlin-induced renal inflammatory responses. The link between gremlin/VEGFR2 and NF-κB/inflammation was confirmed in vitro. Gremlin overexpression was associated with VEGFR2 activation in human renal disease and in the unilateral ureteral obstruction experimental model, where VEGFR2 kinase inhibition diminished renal inflammation. Our data show that a gremlin/VEGFR2 axis participates in renal inflammation and could be a novel target for kidney disease.
Collapse
Affiliation(s)
- Carolina Lavoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | - Matilde Alique
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | - Raquel Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | | | - Gyorgy Keri
- VichemChemie Ltd, Budapest, Hungary.,MTA-SE Pathobiochemistry Research Group, Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Jesús Egido
- Division of Nephrology and Hypertension. IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, CIBERDEM, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| |
Collapse
|
77
|
Iron-hepcidin dysmetabolism, anemia and renal hypoxia, inflammation and fibrosis in the remnant kidney rat model. PLoS One 2015; 10:e0124048. [PMID: 25867633 PMCID: PMC4395008 DOI: 10.1371/journal.pone.0124048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 02/27/2015] [Indexed: 12/15/2022] Open
Abstract
Anemia is a common complication of chronic kidney disease (CKD) that develops early and its severity increases as renal function declines. It is mainly due to a reduced production of erythropoietin (EPO) by the kidneys; however, there are evidences that iron metabolism disturbances increase as CKD progresses. Our aim was to study the mechanisms underlying the development of anemia of CKD, as well as renal damage, in the remnant kidney rat model of CKD induced by 5/6 nephrectomy. This model of CKD presented a sustained degree of renal dysfunction, with mild and advanced glomerular and tubulointerstitial lesions. Anemia developed 3 weeks after nephrectomy and persisted throughout the protocol. The remnant kidney was still able to produce EPO and the liver showed an increased EPO gene expression. In spite of the increased EPO blood levels, anemia persisted and was linked to low serum iron and transferrin levels, while serum interleukin (IL)-6 and high sensitivity C-reactive protein (hs-CRP) levels showed the absence of systemic inflammation. The increased expression of duodenal ferroportin favours iron absorption; however, serum iron is reduced which might be due to iron leakage through advanced kidney lesions, as showed by tubular iron accumulation. Our data suggest that the persistence of anemia may result from disturbances in iron metabolism and by an altered activity/function of EPO as a result of kidney cell damage and a local inflammatory milieu, as showed by the increased gene expression of different inflammatory proteins in the remnant kidney. In addition, this anemia and the associated kidney hypoxia favour the development of fibrosis, angiogenesis and inflammation that may underlie a resistance to EPO stimuli and reduced iron availability. These findings might contribute to open new windows to identify putative therapeutic targets for this condition, as well as for recombinant human EPO (rHuEPO) resistance, which occurs in a considerable percentage of CKD patients.
Collapse
|
78
|
Pallet N, Thervet E, Timsit MO. Angiogenic response following renal ischemia reperfusion injury: new players. Prog Urol 2015; 24 Suppl 1:S20-5. [PMID: 24950928 DOI: 10.1016/s1166-7087(14)70059-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ischemia-reperfusion (IR) injury can negatively influence the short- and long-term outcomes of kidney transplantation because it promotes acute tubular necrosis and tissue scarring and activates innate alloimmunity. The adaptive responses to IR are centrally involved in reducing tissue damage but can also be deleterious when they activate programmed cell death and inflammation. The HIF-1α-mediated angiogenic responses following IR at early and late stages are complex and poorly understood. The early stages of IR seem to be associated with an antiangiogenic response, whereas the hypoxia that follows IR at later stages may activate angiogenic factors such as vascular endothelial growth factor (VEGF) and may be beneficial by stabilizing the microvasculature and favoring local blood supply. In addition to HIF-1α, new players in angiogenesis, including mTOR and the unfolded protein response, may lead to innovative therapeutic strategies for treating patients with ischemia- and reperfusion-associated tissue inflammation and organ dysfunction.
Collapse
Affiliation(s)
- N Pallet
- Service de Biochimie, hôpital européen Georges Pompidou, Paris, France; Université Paris-Descartes, Sorbonne Paris Cité, Paris, France; INSERM U775, Centre universitaire des Saints-Pères, Paris, France.
| | - E Thervet
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France; Service de Néphrologie, hôpital européen Georges Pompidou, université Paris-Descartes, Paris, France
| | - M-O Timsit
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France; Service d'Urologie, hôpital européen Georges-Pompidou, université Paris-Descartes, Paris, France
| |
Collapse
|
79
|
Zhang Z, Zhang F, Lu Y, Zheng S. Update on implications and mechanisms of angiogenesis in liver fibrosis. Hepatol Res 2015; 45:162-78. [PMID: 25196587 DOI: 10.1111/hepr.12415] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/15/2014] [Accepted: 08/31/2014] [Indexed: 02/06/2023]
Abstract
Liver fibrosis occurs as a compensatory response to the process of tissue repair in a wide range of chronic liver injures. It is characterized by excessive deposition of extracellular matrix in liver tissues. As the pathogenesis progresses without effective management, it will lead to formation of liver fiber nodules and disruption of normal liver structure and function, finally culminating in cirrhosis and hepatocellular carcinoma. A new discovery shows that liver angiogenesis is strictly associated with, and may even favor fibrogenic progression of chronic liver diseases. Recent basic and clinical investigations also demonstrate that liver fibrogenesis is accompanied by pathological angiogenesis and sinusoidal remodeling, which critically determine the pathogenesis and prognosis of liver fibrosis. Inhibition of pathological angiogenesis is considered to be a new strategy for the treatment of liver fibrosis. This review summarizes current knowledge on the process of angiogenesis, the relationships between angiogenesis and liver fibrosis, and on the molecular mechanisms of liver angiogenesis. On the other hand, it also presents the different strategies that have been used in experimental models to counteract excessive angiogenesis and the role of angiogenesis in the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Zili Zhang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | | | | | | |
Collapse
|
80
|
Neuwirt H, Perco P, Kainz A, Mühlberger I, Leierer J, Braniff SJ, Mayer B, Mayer G, Rudnicki M. A 3-biomarker-panel predicts renal outcome in patients with proteinuric renal diseases. BMC Med Genomics 2014; 7:75. [PMID: 25540021 PMCID: PMC4301948 DOI: 10.1186/s12920-014-0075-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 12/17/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Clinical and histological parameters are valid prognostic markers in renal disease, although they may show considerable interindividual variability and sometimes limited prognostic value. Novel molecular markers and pathways have the potential to increase the predictive prognostic value of the so called "traditional markers". METHODS Transcriptomics profiles from laser-capture microdissected proximal tubular epithelial cells from routine kidney biopsies were correlated with a chronic renal damage index score (CREDI), an inflammation score (INSCO), and clinical parameters. We used data from 20 renal biopsies with various proteinuric renal diseases with a median follow-up of 49 months (discovery cohort). For validation we performed microarrays from whole kidney biopsies from a second cohort consisting of 16 patients with a median follow-up time of 28 months (validation cohort). RESULTS 562 genes correlated with the CREDI score and 285 genes correlated with the INSCO panel, respectively. 39 CREDI and 90 INSCO genes also correlated with serum creatinine at follow-up. After hierarchical clustering we identified 5 genes from the CREDI panel, and 10 genes from the INSCO panel, respectively, which showed kidney specific gene expression. After exclusion of genes, which correlated to each other by > 50% we identified VEGF-C from the CREDI panel and BMP7, THBS1, and TRIB1 from the INSCO panel. Traditional markers for chronic kidney disease progression and inflammation score predicted 44% of the serum creatinine variation at follow-up. VEGF-C did not further enhance the predictive value, but BMP7, THBS1 and TRIB1 together predicted 94% of the serum creatinine at follow up (p < 0.0001). The model was validated in a second cohort of patients yielding also a significant prediction of follow up creatinine (48%, p = 0.0115). CONCLUSION We identified and validated a panel of three genes in kidney biopsies which predicted serum creatinine at follow-up and therefore might serve as biomarkers for kidney disease progression.
Collapse
|
81
|
Zhao N, Xu Q, Wang M, Fei X, Pan Y, Chen X, Ma S. Mechanism of kidney injury caused by bevacizumab in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8675-83. [PMID: 25674232 PMCID: PMC4314018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/01/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVE We investigate kidney injury caused by high dose bevacizumab to uncover the possible mechanisms involving in this process. METHODS Forty rats were divided into four groups: cisplation group (treated with 1 mg/kg cisplation), Bev-high group (treated with 5 mg/kg bevacizumab); Bev-low group (treated with 2.5 mg/kg bevacizumab) and control group (treated with saline). The urine microalbumin, serum cystatin C, blood urea nitrogen and serum creatinine were detected in the four group rats, respectively. The immunoglobulin of IgG, IgA and IgM and protein of VEGF (vascular endothelial growth factor) and nephrin were detected by immunohistochemical methods. RESULTS All the levels of microalbumin, cystatin C, serum creatinine and blood urea nitrogen in Bev-high group were significantly higher than those in normal control group (P < 0.05). The cystatin C was much more increased in kidney Bev-high group than cisplatin and Bev-low groups (P < 0.05). The light microscope showed a normal glomerular morphology in the four groups, while the electronic microscopy showed the podocytes were extensively fused in cisplatin group and Bev-high group. The two groups were found IgG and IgM deposition as well. The VEGF in kidney amples were down regulated in high dose bevacizumab group, whereas the nephrin and IgA showed no significant expression changes at all. CONCLUSION Bevacizumab increases the risk of injury in glomerular filtration barrier in a dose dependent model. The injury may not only associate with the rising level of proteinuria but also with podocyte-dependent membrane structures.
Collapse
Affiliation(s)
- Ning Zhao
- Department of Nephrology, Hangzhou First People’s HospitalHangzhou 310006, Zhejiang Province, China
| | - Qunhong Xu
- Department of Nephrology, Hangzhou First People’s HospitalHangzhou 310006, Zhejiang Province, China
| | - Ming Wang
- Department of Nephrology, Hangzhou First People’s HospitalHangzhou 310006, Zhejiang Province, China
| | - Xiao Fei
- Department of Nephrology, Hangzhou First People’s HospitalHangzhou 310006, Zhejiang Province, China
| | - Yuelong Pan
- Department of Oncology, Hangzhou Cancer HospitalHangzhou 310006, Zhejiang Province, China
| | - Xueqing Chen
- Department of Oncology, Hangzhou Cancer HospitalHangzhou 310006, Zhejiang Province, China
| | - Shenglin Ma
- Hangzhou First People’s HospitalHangzhou 310006, Zhejiang Province, China
| |
Collapse
|
82
|
Tanaka S, Tanaka T, Nangaku M. Hypoxia as a key player in the AKI-to-CKD transition. Am J Physiol Renal Physiol 2014; 307:F1187-95. [PMID: 25350978 DOI: 10.1152/ajprenal.00425.2014] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent clinical and animal studies have shown that acute kidney injury (AKI), even if followed by complete recovery of renal function, can eventually result in chronic kidney disease (CKD). Renal hypoxia is emerging as a key player in the pathophysiology of the AKI-to-CKD transition. Capillary rarefaction after AKI episodes induces renal hypoxia, which can in turn profoundly affect tubular epithelial cells, (myo)fibroblasts, and inflammatory cells, culminating in tubulointerstitial fibrosis, i.e., progression to CKD. Damaged tubular epithelial cells that fail to redifferentiate might supply a decreased amount of vascular endothelial growth factor and contribute to capillary rarefaction, thus aggravating hypoxia and forming a vicious cycle. Mounting evidence also shows that epigenetic changes are closely related to renal hypoxia in the pathophysiology of CKD progression. Animal experiments suggest that targeting hypoxia is a promising strategy to block the transition from AKI to CKD. However, the precise mechanisms by which hypoxia induces the AKI-to-CKD transition and by which hypoxia-inducible factor activation can exert a protective effect in this context should be clarified in further studies.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
83
|
Donderski R, Szczepanek J, Domagalski K, Tretyn A, Korenkiewicz J, Marszałek A, Szymański A, Wolski Z, Odrowąż-Sypniewska G, Manitius J. Analysis of relative expression level of VEGF ( vascular endothelial growth factor ), HIF-1α ( hypoxia inducible factor 1α ) and CTGF ( connective tissue growth factor ) genes in chronic glomerulonephritis (CGN) patients. Kidney Blood Press Res 2014; 38:83-91. [PMID: 24577260 DOI: 10.1159/000355754] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Analysis of gene expression in renal tissue is considered to be a diagnostic tool predicting the clinical course of glomerulonephritis. The present study quantified the relative transcript levels of VEGF, CTGF and HIF-1α in renal tissue to establish their relationship with some clinical variables in patients suffering from chronic glomerulonephritis (CGN). METHODS 28 patients (6F and 22M, mean age 51.2±15.0) with CGN were enrolled. Type of CNG recognized by kidney biopsy (histopatological evaluation) was as follows: minimal change disease (MCD)-3pts, IgA nephropathy-5pts, FSGS-3pts, membranous nephropathy-4pts, mesangio-proliferative glomerulonephritis-3pts; MPGN-1pts, lupus nephritis-6pts, granulomatosis with polyangitis-2 pts; hypertensive nephropathy- 3pts. Renal tissue from 3 individuals with normal eGFR and histology was taken as control. Mean clinical follow-up of patients was 12 months after biopsy eGFR and daily urinary protein excretion (DPE) was assessed at the time of biopsy and then in 6 months intervals. Real-time PCR was used to determine relative gene expression. The housekeeping gene GAPDH was used as normalization control. RESULTS At the time of the biopsy relative expression of 3 analyzed genes was diminished in comparison to control. There were statistically significant differences in VEGF gene relative expression level in patients which varied according to eGFR and tendency in patients which varied according to DPE. HIF-alfa and CTGF gene showed only a tendency. CONCLUSIONS Overexpression of the VEGF gene in subjects with DPE>3,5 g may point to insufficient oxygen supply in renal tissue which may result in tubulointerstitial fibrosis with further functional renal impairment and decline of eGFR.
Collapse
Affiliation(s)
- Rafał Donderski
- Department of Nephrology, Hypertension and Internal Medicine, Nicolaus Copernicus University Toruń Bydgoszcz, Poland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Liu S, Soong Y, Seshan SV, Szeto HH. Novel cardiolipin therapeutic protects endothelial mitochondria during renal ischemia and mitigates microvascular rarefaction, inflammation, and fibrosis. Am J Physiol Renal Physiol 2014; 306:F970-80. [PMID: 24553434 DOI: 10.1152/ajprenal.00697.2013] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Microvascular rarefaction, or loss of microvascular density, is increasingly implicated in the progression from acute ischemic kidney injury to chronic kidney disease. Microvascular dropout results in chronic tissue hypoxia, interstitial inflammation, and fibrosis. There is currently no therapeutic intervention for microvascular rarefaction. We hypothesize that capillary dropout begins with ischemic damage to endothelial mitochondria due to cardiolipin peroxidation, resulting in loss of cristae and the failure to regenerate ATP upon reperfusion. SS-31 is a cell-permeable peptide that targets the inner mitochondrial membrane and binds selectively to cardiolipin. It was recently shown to inhibit cardiolipin peroxidation by cytochrome c peroxidase activity, and it has been shown to protect mitochondrial cristae in proximal tubular cells during ischemia, and accelerated ATP recovery upon reperfusion. We found mitochondrial swelling and loss of cristae membranes in endothelial and medullary tubular epithelial cells after 45-min ischemia in the rat. The loss of cristae membranes limited the ability of these cells to regenerate ATP upon reperfusion and led to loss of vascular integrity and to tubular cell swelling. SS-31 prevented mitochondria swelling and protected cristae membranes in both endothelial and epithelial cells. By minimizing endothelial and epithelial cell injury, SS-31 prevented "no-reflow" after ischemia and significantly reduced the loss of peritubular capillaries and cortical arterioles, interstitial inflammation, and fibrosis at 4 wk after ischemia. These results suggest that mitochondria protection represents an upstream target for pharmacological intervention in microvascular rarefaction and fibrosis.
Collapse
Affiliation(s)
- Shaoyi Liu
- Dept. of Pharmacology, Weill Cornell Medical College, 1300 York Ave., New York, NY 10021.
| | | | | | | |
Collapse
|
85
|
Quantitative profiling of the rat heart myoblast secretome reveals differential responses to hypoxia and re-oxygenation stress. J Proteomics 2014; 98:138-49. [DOI: 10.1016/j.jprot.2013.12.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/25/2013] [Accepted: 12/28/2013] [Indexed: 11/18/2022]
|
86
|
Abstract
An intact microcirculation is vital for diffusion of oxygen and nutrients and for removal of toxins of every organ and system in the human body. The functional and/or anatomical loss of microvessels is known as rarefaction, which can compromise the normal organ function and have been suggested as a possible starting point of several diseases. The purpose of this overview is to discuss the potential underlying mechanisms leading to renal microvascular rarefaction, and the potential consequences on renal function and on the progression of renal damage. Although the kidney is a special organ that receives much more blood than its metabolic needs, experimental and clinical evidence indicates that renal microvascular rarefaction is associated to prevalent cardiovascular diseases such as diabetes, hypertension, and atherosclerosis, either as cause or consequence. On the other hand, emerging experimental evidence using progenitor cells or angiogenic cytokines supports the feasibility of therapeutic interventions capable of modifying the progressive nature of microvascular rarefaction in the kidney. This overview will also attempt to discuss the potential renoprotective mechanisms of the therapeutic targeting of the renal microcirculation.
Collapse
Affiliation(s)
- Alejandro R Chade
- The Department of Physiology and Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| |
Collapse
|
87
|
Menzies RI, Zammit-Mangion A, Hollis LM, Lennen RJ, Jansen MA, Webb DJ, Mullins JJ, Dear JW, Sanguinetti G, Bailey MA. An anatomically unbiased approach for analysis of renal BOLD magnetic resonance images. Am J Physiol Renal Physiol 2013; 305:F845-52. [DOI: 10.1152/ajprenal.00113.2013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Oxygenation defects may contribute to renal disease progression, but the chronology of events is difficult to define in vivo without recourse to invasive methodologies. Blood oxygen level-dependent magnetic resonance imaging (BOLD MRI) provides an attractive alternative, but the R2* signal is physiologically complex. Postacquisition data analysis often relies on manual selection of region(s) of interest. This approach excludes from analysis significant quantities of biological information and is subject to selection bias. We present a semiautomated, anatomically unbiased approach to compartmentalize voxels into two quantitatively related clusters. In control F344 rats, low R2* clustering was located predominantly within the cortex and higher R2* clustering within the medulla (70.96 ± 1.48 vs. 79.00 ± 1.50; 3 scans per rat; n = 6; P < 0.01) consistent anatomically with a cortico-medullary oxygen gradient. An intravenous bolus of acetylcholine caused a transient reduction of the R2* signal in both clustered segments ( P < 0.01). This was nitric oxide dependent and temporally distinct from the hemodynamic effects of acetylcholine. Rats were then chronically infused with angiotensin II (60 ng/min) and rescanned 3 days later. Clustering demonstrated a disruption of the cortico-medullary gradient, producing less distinctly segmented mean R2* clusters (71.30 ± 2.00 vs. 72.48 ± 1.27; n = 6; NS). The acetylcholine-induced attenuation of the R2* signal was abolished by chronic angiotensin II infusion, consistent with reduced nitric oxide bioavailability. This global map of oxygenation, defined by clustering individual voxels on the basis of quantitative nearness, might be more robust in defining deficits in renal oxygenation than the absolute magnitude of R2* in small, manually selected regions of interest defined exclusively by anatomical nearness.
Collapse
Affiliation(s)
- Robert I. Menzies
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | | | - Lyam M. Hollis
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | - Ross J. Lennen
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | - Maurits A. Jansen
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | - David J. Webb
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | - John J. Mullins
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | - James W. Dear
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| | - Guido Sanguinetti
- School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew A. Bailey
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom; and
| |
Collapse
|
88
|
Abstract
PURPOSE OF REVIEW Pathological deposition of fibrous matrix in organs is a major problem and contributes to as many as 45% of all natural deaths. Chronic kidney disease affects 8% of the US population, and is characterized by fibrotic processes. It frequently progresses to organ failure and is a major cause of cardiovascular death; yet it lacks therapies. Understanding the pathological mechanisms of fibrosis in the kidney and other organs is central to the development of new therapeutics. RECENT FINDINGS Pericytes are mesenchymal cells that partially cover capillary walls. Pericytes play critical roles in micro-vessel formation, maturation and stability. New genetic fate-mapping studies have identified pericytes and the closely related resident fibroblasts as the major progenitors of scar-forming myofibroblasts in multiple organs including the kidney, appearing in response to tissue injury. When pericytes become myofibroblasts they lose pericyte functions. Capillaries become unstable with deleterious consequences for the kidney. The cellular and molecular mechanisms underpinning these processes are starting to unravel, leading to new therapeutics for chronic fibrosing diseases of the kidney and potentially other organs. SUMMARY This review focuses on pericytes in the kidney and other organs, their role in fibrogenesis and their role in regulation of the microvasculature.
Collapse
|
89
|
Basile DP, Zeng P, Friedrich JL, Leonard EC, Yoder MC. Low proliferative potential and impaired angiogenesis of cultured rat kidney endothelial cells. Microcirculation 2013; 19:598-609. [PMID: 22612333 DOI: 10.1111/j.1549-8719.2012.00193.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE CKD is histologically characterized by interstitial fibrosis, which may be driven by peritubular capillary dropout and hypoxia. Surprisingly, peritubular capillaries have little repair capacity. We sought to establish long-term cultures of rat kidney endothelial cells to investigate their growth regulatory properties. METHODS AKEC or YKEC were isolated using CD31-based isolation techniques and sustained in long-term cultures. RESULTS Although YKEC grew slightly better than AKEC, both performed poorly compared with endothelial cells of the rat adult PMVEC, PAEC, or HUVEC cells. PMVEC and PAEC contained a large percentage of cells with high colony-forming potential. In contrast, KECs were incapable of forming large colonies and most remained as single nondividing cells. KEC expressed high levels of mRNA for VEGF receptors, but were surprisingly insensitive to VEGF stimulation. KEC did not form branching structures on Matrigel when cultured alone, but in mixed cultures, KEC incorporated into branching structures with PMVEC. CONCLUSIONS These data suggest that the intrinsic growth of rat kidney endothelial cells is limited by unknown mechanisms. The low growth rate may be related to the minimal intrinsic regenerative capacity of renal capillaries.
Collapse
Affiliation(s)
- David P Basile
- Department of Cellular and Integrative Physiology, Indiana University, Indianapolis 46202, USA.
| | | | | | | | | |
Collapse
|
90
|
Habenicht LM, Webb TL, Clauss LA, Dow SW, Quimby JM. Urinary cytokine levels in apparently healthy cats and cats with chronic kidney disease. J Feline Med Surg 2013; 15:99-104. [PMID: 22989558 PMCID: PMC10816647 DOI: 10.1177/1098612x12461007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chronic kidney disease (CKD) is a common cause of illness and death in cats. The hallmark of CKD in cats is chronic tubulointerstitial nephritis, and inflammation contributes to the progression of renal fibrosis. However, at present, it is difficult to assess directly the degree of intra-renal inflammation without renal biopsy. Measurement of inflammatory cytokine levels in urine may provide a non-invasive means of assessing intra-renal inflammation. Urine cytokine levels (urine cytokine/urine creatinine ratio) were measured in 18 healthy cats and 26 cats with CKD. When urine cytokine levels in healthy and CKD cats were compared, we found significantly higher levels of IL-8 and transforming growth factor-β1 (TGF-β1) in urine of CKD cats, along with significantly lower vascular endothelial growth factor (VEGF) levels. A significant positive correlation between serum creatinine and TGF-β1 levels was found in CKD cats. Urinary cytokine measurement may, potentially, be a useful means of assessing intra-renal inflammation, fibrosis and vascular health in cats with CKD.
Collapse
Affiliation(s)
- Lauren M Habenicht
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | | | | |
Collapse
|
91
|
Tung-Wei H, Jia-Hung L, Kun-Tu Y, Jen-Pi T, Sheng-Wen W, Hui-Chun T, Wei-Tse K, Shu-Hui L, Ya-Wen C, Horng-Rong C. Renal expression of hypoxia inducible factor-1α in patients with chronic kidney disease: a clinicopathologic study from nephrectomized kidneys. Indian J Med Res 2013; 137:102-10. [PMID: 23481058 PMCID: PMC3657871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND & OBJECTIVES Hypoxia inducible factor-1α (HIF-1α) has been shown to play a role in the pathogenesis of renal interstitial fibrosis. However, the relationship of HIF-1α expression intensity in human renal tissue with the degree of renal function or renal fibrosis has not been investigated. We therefore, undertook this study to assess the relationship between HIF-1α expression and degree of renal impairment and renal fibrosis using renal tissue from nephrectomized kidneys from patients with chronic kidney disease. METHODS This retrospective study was performed with 70 patients undergoing unilateral or bilateral nephrectomy because of renal cell carcinoma, urothelial cell carcinoma, or renal abscess. Immunohistochemical analysis of HIF-1α expression in non-tumourous or non-abscess renal parenchyma was performed. The patients were divided into two groups: group 1 (n=37) with low intensity HIF-1α expression and group 2 (n=33) with high intensity HIF-1α expression. RESULTS The intensity of renal HIF-1α expression was significantly associated with serum creatinine level (P =0.005), estimated glomerular filtration rate (P=0.02), fibrosis score of the interstitium (P=0.004) and glomerular sclerosis (P=0.013). A high intensity of HIF-1α expression tended to be associated with lower serum creatinine, higher estimated glomerular filtration rate, low interstitial fibrosis score and low glomerular sclerosis. In addition, multivariate analysis by step-wise logistic regression demonstrated that interstitial fibrosis was the only independent factor associated with the intensity of renal HIF-1α expression (OR 4.107, CI 1.535-11.313, P=0.005). INTERPRETATION & CONCLUSIONS This study demonstrated a correlation between intensity of HIF-1α expression and degree of renal interstitial fibrosis. The association demonstrated an elevated HIF-1α expression in less severe kidney disease. The intensity of HIF-1α renal expression plays a role in the pathogenesis of chronic kidney disease.
Collapse
Affiliation(s)
- Hung Tung-Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Division of Nephrology, Department of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Liou Jia-Hung
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Yeh Kun-Tu
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tsai Jen-Pi
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Departments of Nephrology, Buddhist Dalin Tzu Chi General Hospital, Dalin, Taiwan
| | - Wu Sheng-Wen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Division of Nephrology, Department of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tai Hui-Chun
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Kao Wei-Tse
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Lin Shu-Hui
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan,Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing & Management, Miaoli, Taiwan
| | - Cheng Ya-Wen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chang Horng-Rong
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Division of Nephrology, Department of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
92
|
Dormond O, Dufour M, Seto T, Bruneau S, Briscoe DM. Targeting the intragraft microenvironment and the development of chronic allograft rejection. Hum Immunol 2012; 73:1261-8. [PMID: 22863981 DOI: 10.1016/j.humimm.2012.07.334] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 06/29/2012] [Accepted: 07/25/2012] [Indexed: 01/10/2023]
Abstract
In this review, we discuss a paradigm whereby changes in the intragraft microenvironment promote or sustain the development of chronic allograft rejection. A key feature of this model involves the microvasculature including (a) endothelial cell (EC) destruction, and (b) EC proliferation, both of which result from alloimmune leukocyte- and/or alloantibody-induced responses. These changes in the microvasculature likely create abnormal blood flow patterns and thus promote local tissue hypoxia. Another feature of the chronic rejection microenvironment involves the overexpression of vascular endothelial growth factor (VEGF). VEGF stimulates EC activation and proliferation and it has potential to sustain inflammation via direct interactions with leukocytes. In this manner, VEGF may promote ongoing tissue injury. Finally, we review how these events can be targeted therapeutically using mTOR inhibitors. EC activation and proliferation as well as VEGF-VEGFR interactions require PI-3K/Akt/mTOR intracellular signaling. Thus, agents that inhibit this signaling pathway within the graft may also target the progression of chronic rejection and thus promote long-term graft survival.
Collapse
Affiliation(s)
- Olivier Dormond
- The Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
93
|
Ebrahimi B, Li Z, Eirin A, Zhu XY, Textor SC, Lerman LO. Addition of endothelial progenitor cells to renal revascularization restores medullary tubular oxygen consumption in swine renal artery stenosis. Am J Physiol Renal Physiol 2012; 302:F1478-85. [PMID: 22419692 PMCID: PMC3378175 DOI: 10.1152/ajprenal.00563.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/08/2012] [Indexed: 01/12/2023] Open
Abstract
Renal artery stenosis (RAS) promotes microvascular rarefaction and fibrogenesis, which may eventuate in irreversible kidney injury. We have shown that percutaneous transluminal renal angioplasty (PTRA) or endothelial progenitor cells (EPC) improve renal cortical hemodynamics and function in the poststenotic kidney. The renal medulla is particularly sensitive to hypoxia, yet little is known about reversibility of medullary injury on restoration of renal blood flow. This study was designed to test the hypothesis that PTRA, with or without adjunct EPC delivery to the stenotic kidney, may improve medullary remodeling and tubular function. RAS was induced in 21 pigs using implantation of irritant coils, while another group served as normal controls (n = 7 each). Two RAS groups were then treated 6 wk later with PTRA or both PTRA and EPC. Four weeks later, medullary hemodynamics, microvascular architecture, and oxygen-dependent tubular function of the stenotic kidneys were examined using multidetector computed tomography, microcomputed tomography, and blood oxygenation level-dependent MRI, respectively. Medullary protein expression of vascular endothelial growth factor, endothelial nitric oxide synthase, hypoxia-inducible factor-1α, and NAD(P)H oxidase p47 were determined. All RAS groups showed decreased medullary vascular density and blood flow. However, in RAS+PTRA+EPC animals, EPC were engrafted in tubular structures, oxygen-dependent tubular function was normalized, and fibrosis attenuated, despite elevated expression of hypoxia-inducible factor-1α and sustained downregulation of vascular endothelial growth factor. In conclusion, EPC delivery, in addition to PTRA, restores medullary oxygen-dependent tubular function, despite impaired medullary blood and oxygen supply. These results support further development of cell-based therapy as an adjunct to revascularization of RAS.
Collapse
Affiliation(s)
- Behzad Ebrahimi
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
94
|
Abstract
Pericytes are cells of mesenchymal origin that are intimately involved in the development and stabilization of vascular networks. Novel studies of their role in inflammation have identified that pericytes are not only major contributors to the activated matrix depositing myofibroblast populations seen in progressive renal fibrosis but perhaps even more importantly, the detachment of renal pericytes from the vasculature contributes to the microvasculature rarefaction and subsequent hypoxia associated with chronic kidney disease. In this review, our current understanding of the functioning of renal pericytes will be considered and set in the context of the wider literature that is currently available on this neglected population of cells.
Collapse
|
95
|
Bouvier N, Fougeray S, Beaune P, Thervet E, Pallet N. The unfolded protein response regulates an angiogenic response by the kidney epithelium during ischemic stress. J Biol Chem 2012; 287:14557-68. [PMID: 22403402 PMCID: PMC3340272 DOI: 10.1074/jbc.m112.340570] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 03/02/2012] [Indexed: 12/20/2022] Open
Abstract
Ischemic injuries permanently affect kidney tissue and challenge cell viability, promoting inflammation and fibrogenesis. Ischemia results in nutrient deprivation, which triggers endoplasmic reticulum stress, ultimately resulting in the unfolded protein response (UPR). The aim of this study was to test whether the UPR could promote an angiogenic response independently of the HIF-1α pathway during ischemic stress in the human kidney epithelium. Glucose deprivation induced the secretion of vascular endothelial growth factor A (VEGFA), basic fibroblast growth factor (bFGF) and angiogenin (ANG) in human kidney epithelial cells independently of HIF-1α. Glucose deprivation, but not hypoxia, triggered endoplasmic reticulum stress and activated the UPR. RNA interference-mediated inhibition of the gene encoding the kinase PERK decreased VEGFA and bFGF expression, but neither gene was affected by the inhibition of IRE1α or ATF6. Furthermore, we show that the expression of angiogenin, which inhibits protein synthesis, is regulated by both IRE1α and PERK, which could constitute a complementary function of the UPR in the repression of translation. In a rat model of acute ischemic stress, we show that the UPR is activated in parallel with VEGFA, bFGF, and ANG expression and independently of HIF-1α.
Collapse
Affiliation(s)
- Nicolas Bouvier
- From INSERM U775, Centre Universitaire des Saints Peres, Paris, France
- the Universite Paris Descartes, Sorbonne Paris Cite, Paris 75065, France
| | - Sophie Fougeray
- From INSERM U775, Centre Universitaire des Saints Peres, Paris, France
- the Universite Paris Descartes, Sorbonne Paris Cite, Paris 75065, France
| | - Philippe Beaune
- From INSERM U775, Centre Universitaire des Saints Peres, Paris, France
- Pole de Biologie, Hopital Europeen Georges Pompidou, Paris 70115, France, and
- the Universite Paris Descartes, Sorbonne Paris Cite, Paris 75065, France
| | - Eric Thervet
- From INSERM U775, Centre Universitaire des Saints Peres, Paris, France
- the Service de Nephrologie, and
- the Universite Paris Descartes, Sorbonne Paris Cite, Paris 75065, France
| | - Nicolas Pallet
- From INSERM U775, Centre Universitaire des Saints Peres, Paris, France
- the Universite Paris Descartes, Sorbonne Paris Cite, Paris 75065, France
| |
Collapse
|
96
|
Bruneau S, Woda CB, Daly KP, Boneschansker L, Jain NG, Kochupurakkal N, Contreras AG, Seto T, Briscoe DM. Key Features of the Intragraft Microenvironment that Determine Long-Term Survival Following Transplantation. Front Immunol 2012; 3:54. [PMID: 22566935 PMCID: PMC3342046 DOI: 10.3389/fimmu.2012.00054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/02/2012] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss how changes in the intragraft microenvironment serve to promote or sustain the development of chronic allograft rejection. We propose two key elements within the microenvironment that contribute to the rejection process. The first is endothelial cell proliferation and angiogenesis that serve to create abnormal microvascular blood flow patterns as well as local tissue hypoxia, and precedes endothelial-to-mesenchymal transition. The second is the overexpression of local cytokines and growth factors that serve to sustain inflammation and, in turn, function to promote a leukocyte-induced angiogenesis reaction. Central to both events is overexpression of vascular endothelial growth factor (VEGF), which is both pro-inflammatory and pro-angiogenic, and thus drives progression of the chronic rejection microenvironment. In our discussion, we focus on how inflammation results in angiogenesis and how leukocyte-induced angiogenesis is pathological. We also discuss how VEGF is a master control factor that fosters the development of the chronic rejection microenvironment. Overall, this review provides insight into the intragraft microenvironment as an important paradigm for future direction in the field.
Collapse
Affiliation(s)
- Sarah Bruneau
- The Division of Nephrology, Transplantation Research Center, Children's Hospital Boston Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Abstract
Chronic kidney disease is characterized by progressive loss of the renal microvasculature, which leads to local areas of hypoxia and induction of profibrotic responses, scarring and deterioration of renal function. Revascularization alone might be sufficient to restore kidney function and regenerate the structure of the diseased kidney. For revascularization to be successful, however, the underlying disease process needs to be halted or alleviated and there must remain a sufficient number of surviving nephron units that can serve as a scaffold for kidney regeneration. This Perspectives article describes how revascularization might be achieved using vascular growth factors or adoptive transfer of endothelial progenitor cells and provides a brief outline of the studies performed to date. An overview of how therapeutic strategies targeting the microvasculature could be enhanced in the future is also presented.
Collapse
|