1
|
Pan W, Zhang W, Zheng B, Camellato BR, Stern J, Lin Z, Khodadadi-Jamayran A, Kim J, Sommer P, Khalil K, Weldon E, Bai J, Zhu Y, Meyn P, Heguy A, Mangiola M, Griesemer A, Keating BJ, Montgomery RA, Xia B, Boeke JD. Cellular dynamics in pig-to-human kidney xenotransplantation. MED 2024; 5:1016-1029.e4. [PMID: 38776915 DOI: 10.1016/j.medj.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/30/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Xenotransplantation of genetically engineered porcine organs has the potential to address the challenge of organ donor shortage. Two cases of porcine-to-human kidney xenotransplantation were performed, yet the physiological effects on the xenografts and the recipients' immune responses remain largely uncharacterized. METHODS We performed single-cell RNA sequencing (scRNA-seq) and longitudinal RNA-seq analyses of the porcine kidneys to dissect xenotransplantation-associated cellular dynamics and xenograft-recipient interactions. We additionally performed longitudinal scRNA-seq of the peripheral blood mononuclear cells (PBMCs) to detect recipient immune responses across time. FINDINGS Although no hyperacute rejection signals were detected, scRNA-seq analyses of the xenografts found evidence of endothelial cell and immune response activation, indicating early signs of antibody-mediated rejection. Tracing the cells' species origin, we found human immune cell infiltration in both xenografts. Human transcripts in the longitudinal bulk RNA-seq revealed that human immune cell infiltration and the activation of interferon-gamma-induced chemokine expression occurred by 12 and 48 h post-xenotransplantation, respectively. Concordantly, longitudinal scRNA-seq of PBMCs also revealed two phases of the recipients' immune responses at 12 and 48-53 h. Lastly, we observed global expression signatures of xenotransplantation-associated kidney tissue damage in the xenografts. Surprisingly, we detected a rapid increase of proliferative cells in both xenografts, indicating the activation of the porcine tissue repair program. CONCLUSIONS Longitudinal and single-cell transcriptomic analyses of porcine kidneys and the recipient's PBMCs revealed time-resolved cellular dynamics of xenograft-recipient interactions during xenotransplantation. These cues can be leveraged for designing gene edits and immunosuppression regimens to optimize xenotransplantation outcomes. FUNDING This work was supported by NIH RM1HG009491 and DP5OD033430.
Collapse
Affiliation(s)
- Wanqing Pan
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Binghan Zheng
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brendan R Camellato
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ziyan Lin
- Applied Bioinformatics Laboratories (ABL), NYU Grossman School of Medicine, New York, NY 10016, USA
| | | | - Jacqueline Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Philip Sommer
- Department of Anesthesiology, Perioperative Care & Pain Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jiangshan Bai
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yinan Zhu
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Peter Meyn
- Genome Technology Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Heguy
- Genome Technology Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA; Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA.
| | - Bo Xia
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Society of Fellows, Harvard University, Cambridge, MA 02138, USA.
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
2
|
Liu H, Huang L, Chen F, Zhong Z, Ma X, Zhou Z, Cao S, Shen L, Peng G. Adipose-derived mesenchymal stem cells secrete extracellular vesicles: A potential cell-free therapy for canine renal ischaemia-reperfusion injury. Vet Med Sci 2023; 9:1134-1142. [PMID: 36913179 DOI: 10.1002/vms3.1105] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/15/2023] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stem cells (ADMSCs) and their extracellular vesicles (EVs) are a promising source of therapies for ischaemia-reperfusion (IR) because of their potent anti-inflammatory and immunomodulatory properties. OBJECTIVES The aims of this study were to explore the therapeutic efficacy and potential mechanism of ADMSC-EVs in canine renal IR injury. METHODS Mesenchymal stem cells (MSCs) and EVs were isolated and characterised for surface markers. A canine IR model administered with ADMSC-EVs was used to evaluate therapeutic effects on inflammation, oxidative stress, mitochondrial damage and apoptosis. RESULTS CD105, CD90 and beta integrin ITGB were positively expressed in MSCs, while CD63, CD9 and intramembrane marker TSG101 were positively expressed in EVs. Compared with the IR model group, there was less mitochondrial damage and reduction in quantity of mitochondria in the EV treatment group. Renal IR injury led to severe histopathological lesions and significant increases in biomarkers of renal function, inflammation and apoptosis, which were attenuated by the administration of ADMSC-EVs. CONCLUSIONS Secretion of EVs by ADMSCs exhibited therapeutic potential in renal IR injury and may lead to a cell-free therapy for canine renal IR injury. These findings revealed that canine ADMSC-EVs potently attenuate renal IR injury-induced renal dysfunction, inflammation and apoptosis, possibly by reducing mitochondrial damage.
Collapse
Affiliation(s)
- Haifeng Liu
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liyuan Huang
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Fuhao Chen
- Chongqing Fengdu Agricultural Science and Technology Development Group Co. Ltd, Chongqing, China
| | - Zhijun Zhong
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoping Ma
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ziyao Zhou
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Suizhong Cao
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liuhong Shen
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guangneng Peng
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
3
|
Lin L, Hu K. Annexin A2 and Kidney Diseases. Front Cell Dev Biol 2022; 10:974381. [PMID: 36120574 PMCID: PMC9478026 DOI: 10.3389/fcell.2022.974381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Annexin A2 is a Ca2+- and phospholipid-binding protein which is widely expressed in various types of cells and tissues. As a multifunctional molecule, annexin A2 is found to be involved in diverse cell functions and processes, such as cell exocytosis, endocytosis, migration and proliferation. As a receptor of plasminogen and tissue plasminogen activator, annexin A2 promotes plasmin generation and regulates the homeostasis of blood coagulation, fibrinolysis and matrix degradation. As an antigen expressed on cell membranes, annexin A2 initiates local inflammation and damage through binding to auto-antibodies. Annexin A2 also mediates multiple signaling pathways induced by various growth factors and oxidative stress. Aberrant expression of annexin A2 has been found in numerous kidney diseases. Annexin A2 has been shown to act as a co-receptor of integrin CD11b mediating NF-kB-dependent kidney inflammation, which is further amplified through annexin A2/NF-kB-triggered macrophage M2 to M1 phenotypic change. It also modulates podocyte cytoskeleton rearrangement through Cdc42 and Rac1/2/3 Rho pathway causing proteinuria. Thus, annexin A2 is implicated in the pathogenesis and progression of various kidney diseases. In this review, we focus on the current understanding of the role of annexin A2 in kidney diseases.
Collapse
Affiliation(s)
- Ling Lin
- *Correspondence: Ling Lin, ; Kebin Hu,
| | - Kebin Hu
- *Correspondence: Ling Lin, ; Kebin Hu,
| |
Collapse
|
4
|
Moreno JCA, Bahmad HF, Febres-Aldana CA, Pirela A, Azuero A, Salami A, Poppiti R. Post-mortem assessment of vimentin expression as a biomarker for renal tubular regeneration following acute kidney injury. J Pathol Transl Med 2021; 55:369-379. [PMID: 34638220 PMCID: PMC8601956 DOI: 10.4132/jptm.2021.08.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common cause of morbidity and mortality. It mainly targets the renal tubular epithelium with pathological changes, referred to as acute tubular injury. The latter is followed by a regenerative response that is difficult to visualize on routine hematoxylin and eosin (H&E) stains. In this study, we examined the regenerative capacity of renal tubules by correlating vimentin (VIM) immunohistochemical (IHC) expression and pathological findings of AKI and renal tubular regeneration (RTR) on H&E. METHODS We reviewed 23 autopsies performed in the clinical setting of AKI and RTR. VIM expression was scored in the renal cortical tubular epithelium using a statistical cutoff ≥ 3% for high expression and < 3% for low expression. RESULTS Of the 23 kidney tissues examined, seven (30.4%) had low VIM expression, and 16 (69.6%) had high VIM expression. Kidney tissues with evidence of AKI and RTR had significantly higher VIM expression. Renal peritubular microenvironment features showing regenerative changes on H&E were associated with high VIM expression. In the univariate model, kidney tissues with RTR were 18-fold more likely to have high VIM expression. CONCLUSIONS In conclusion, our findings suggest that VIM could serve as an IHC marker for RTR following AKI. However, correlation with H&E findings remains critical to excluding chronic tubular damage. Collectively, our preliminary results pave the way for future studies including a larger sample size to validate the use of VIM as a reliable biomarker for RTR.
Collapse
Affiliation(s)
- Juan Carlos Alvarez Moreno
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Christopher A Febres-Aldana
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Andrés Pirela
- Department of Internal Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Andres Azuero
- Department of Internal Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Ali Salami
- Department of Mathematics, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Robert Poppiti
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
5
|
Spencer S, Wheeler-Jones C, Elliott J. Hypoxia and chronic kidney disease: Possible mechanisms, therapeutic targets, and relevance to cats. Vet J 2021; 274:105714. [PMID: 34252550 DOI: 10.1016/j.tvjl.2021.105714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
There is mounting evidence that kidney ischaemia/hypoxia plays an important role in feline chronic kidney disease (CKD) development and progression, as well as in human disease and laboratory animal models. Ischaemic acute kidney injury is widely accepted as a cause of CKD in people and data from laboratory species has identified some of the pathways underlying this continuum. Experimental kidney ischaemia in cats results in morphological changes, namely chronic tubulointerstitial inflammation, tubulointerstitial fibrosis, and tubular atrophy, akin to those observed in naturally-occurring CKD. Multiple situations are envisaged that could result in acute or chronic episodes of kidney hypoxia in cats, while risk factors identified in epidemiological studies provide further support that kidney hypoxia contributes to spontaneously occurring feline CKD. This review evaluates the evidence for the role of kidney ischaemia/hypoxia in feline CKD and the proposed mechanisms and consequences of kidney hypoxia. As no effective treatments exist that substantially slow or prevent feline CKD progression, there is a need for novel therapeutic strategies. Targeting kidney hypoxia is one such promising approach, with therapies including those that attenuate the hypoxia-inducible factor (HIF) pathway already being utilised in human CKD.
Collapse
Affiliation(s)
- Sarah Spencer
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| | - Caroline Wheeler-Jones
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Jonathan Elliott
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| |
Collapse
|
6
|
Romero-Palomo F, Festag M, Lenz B, Schadt S, Brink A, Kipar A, Steinhuber B, Husser C, Koller E, Sewing S, Tessier Y, Dzygiel P, Fischer G, Winter M, Hetzel U, Mihatsch MJ, Braendli-Baiocco A. Safety, Tissue Distribution, and Metabolism of LNA-Containing Antisense Oligonucleotides in Rats. Toxicol Pathol 2021; 49:1174-1192. [PMID: 34060347 DOI: 10.1177/01926233211011615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Antisense oligonucleotides (ASOs) are chemically modified nucleic acids with therapeutic potential, some of which have been approved for marketing. We performed a study in rats to investigate mechanisms of toxicity after administration of 3 tool locked nucleic acid (LNA)-containing ASOs with differing established safety profiles. Four male rats per group were dosed once, 3, or 6 times subcutaneously, with 7 days between dosing, and sacrificed 3 days after the last dose. These ASOs were either unconjugated (naked) or conjugated with N-acetylgalactosamine for hepatocyte-targeted delivery. The main readouts were in-life monitoring, clinical and anatomic pathology, exposure assessment and metabolite identification in liver and kidney by liquid chromatography coupled to tandem mass spectrometry, ASO detection in liver and kidney by immunohistochemistry, in situ hybridization, immune electron microscopy, and matrix-assisted laser desorption/ionization mass spectrometry imaging. The highly toxic compounds showed the greatest amount of metabolites and a low degree of tissue accumulation. This study reveals different patterns of cell death associated with toxicity in liver (apoptosis and necrosis) and kidney (necrosis only) and provides new ultrastructural insights on the tissue accumulation of ASOs. We observed that the immunostimulatory properties of ASOs can be either primary from sequence-dependent properties or secondary to cell necrosis.
Collapse
Affiliation(s)
- Fernando Romero-Palomo
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Matthias Festag
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Barbara Lenz
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Simone Schadt
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Andreas Brink
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, 30843Vetsuisse Faculty, University of Zürich, Switzerland
| | - Bernd Steinhuber
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Christophe Husser
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Erich Koller
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Sabine Sewing
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Yann Tessier
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Pawel Dzygiel
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Guy Fischer
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Michael Winter
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| | - Udo Hetzel
- Electron Microscopy Unit, Institute of Veterinary Pathology, 27217Vetsuisse Faculty, University of Zürich, Switzerland
| | - Michael J Mihatsch
- 361703Institute for Pathology, University Hospital of Basel, Switzerland
| | - Annamaria Braendli-Baiocco
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, 1529Roche Innovation Center Basel, Switzerland
| |
Collapse
|
7
|
Abstract
Immunoglobulin A nephropathy (IgAN) is the most common primary glomerular disease all over the world and it is a major cause of renal failure. IgAN prediction in children with machine learning algorithms has been rarely studied. We retrospectively analyzed the electronic medical records from the Nanjing Eastern War Zone Hospital, chose eXtreme Gradient Boosting (XGBoost), random forest (RF), CatBoost, support vector machines (SVM), k-nearest neighbor (KNN), and extreme learning machine (ELM) models in order to predict the probability that the patient would not reach or reach end-stage renal disease (ESRD) within five years, used the chi-square test to select the most relevant 16 features as the input of the model, and designed a decision-making system (DMS) of IgAN prediction in children that is based on XGBoost and Django framework. The receiver operating characteristic (ROC) curve was used in order to evaluate the performance of the models and XGBoost had the best performance by comparison. The AUC value, accuracy, precision, recall, and f1-score of XGBoost were 85.11%, 78.60%, 75.96%, 76.70%, and 76.33%, respectively. The XGBoost model is useful for physicians and pediatric patients in providing predictions regarding IgAN. As an advantage, a DMS can be designed based on the XGBoost model to assist a physician to effectively treat IgAN in children for preventing deterioration.
Collapse
|
8
|
Wen Y, Yang C, Menez SP, Rosenberg AZ, Parikh CR. A Systematic Review of Clinical Characteristics and Histologic Descriptions of Acute Tubular Injury. Kidney Int Rep 2020; 5:1993-2001. [PMID: 33163720 PMCID: PMC7609907 DOI: 10.1016/j.ekir.2020.08.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/13/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The term "acute tubular injury" (ATI) represents histopathologic renal tubular injury and often manifests clinically as acute kidney injury (AKI). Studies systematically summarizing the clinical presentation and histological changes in human ATI are limited. METHODS We used a comprehensive search strategy to search human studies of ATI from 1936 to July 2019. We extracted study characteristics, clinical characteristics, and histologic descriptions of ATI by bright field, immunofluorescence, electron microscopy, and immunohistochemistry. We compared ATI histology as a function of tissue procurement type, timing, and etiologies. RESULTS We included 292 studies comprising a total of 1987 patients. The majority of studies (222 of 292, 76%) were single-center case reports. The mean age of included patients was 47 years. In native kidney biopsy cases, baseline, peak, and latest creatinine were 1.3 mg/dl, 7.19 mg/dl, and 1.85 mg/dl respectively, and biopsy was performed mostly after peak creatinine (86.7%, 391 of 451). We identified 16 histologic descriptions of tubular injury, including tubular cell sloughing (115 of 292, 39.4%), tubular epithelial flattening/simplification (110 of 292, 37.7%), tubular dilatation (109 of 292, 37.3%), and tubular cell necrosis (93 of 292, 31.8%). There was no difference in tubular injury histology among different tissue procurement types (native kidney biopsy, transplant kidney biopsy, and autopsy), among different etiologies, or between different tissue procurement timing (before or after creatinine peaks in native kidneys). Electron microscopy and immunohistochemistry were used in a minority of studies. CONCLUSION ATI manifests with diverse histologic changes. Efforts to establish protocols to harmonize biopsy practices, to handle kidney biopsy for tissue interrogation, and to report results across clinical practice are needed to improve our understanding of this complex disease.
Collapse
Affiliation(s)
- Yumeng Wen
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Yang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Steven P. Menez
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Correspondence: Chirag R. Parikh, Division of Nephrology, Johns Hopkins University School of Medicine, 1830 E. Monument St., Suite 416, Baltimore, Maryland 21287, USA.
| |
Collapse
|
9
|
Abstract
BACKGROUND The potential of a mesenchymal stem cell (MSC) therapy to accelerate the repair of ischemically damaged human kidneys during 24 hours of warm perfusion was evaluated. The hypothesis was that by administering MSC directly to the renal tissue, there would be an improved opportunity for cellular repair mediated by intrarenal paracrine effects. METHODS Studies were performed using the exsanguinous metabolic support (EMS) tissue-engineering platform. Five pairs of human kidney allografts from donation after circulatory death donors were studied. One human kidney was EMS perfused for 24 hours (control), whereas its paired kidney was EMS perfused with MSC (1 × 10). The kidneys were evaluated for DNA synthesis, cytokine/chemokine synthesis, cytoskeletal regeneration, and mitosis. RESULTS Treatment with MSC resulted in reduced inflammatory cytokines synthesized by the kidneys. Mesenchymal stem cell treatment led to a significant increase in the synthesis of adenosine triphosphate and growth factors resulting in normalization of metabolism and the cytoskeleton. Toluidine Blue staining of MSC-treated kidneys demonstrated a significant increase in the number of renal cells undergoing mitosis (26%) compared with EMS perfusion alone. CONCLUSIONS To our knowledge, our work is the first to have demonstrated actual renal regeneration while ischemically damaged human kidneys are perfused ex vivo for 24 hours. The observed regeneration entails: increased synthesis of adenosine triphosphate, a reduced inflammatory response, increased synthesis of growth factors, normalization of the cytoskeleton and mitosis. The ability to regenerate renal tissue ex vivo sufficiently to result in immediate function could revolutionize transplantation by solving the chronic organ shortage.
Collapse
|
10
|
Aristolochic Acids: Newly Identified Exposure Pathways of this Class of Environmental and Food-Borne Contaminants and its Potential Link to Chronic Kidney Diseases. TOXICS 2019; 7:toxics7010014. [PMID: 30893813 PMCID: PMC6468885 DOI: 10.3390/toxics7010014] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 11/17/2022]
Abstract
Aristolochic acids (AAs) are nitrophenanthrene carboxylic acids naturally produced by Aristolochia plants. These plants were widely used to prepare herbal remedies until AAs were observed to be highly nephrotoxic and carcinogenic to humans. Although the use of AA-containing Aristolochia plants in herbal medicine is prohibited in countries worldwide, emerging evidence nevertheless has indicated that AAs are the causative agents of Balkan endemic nephropathy (BEN), an environmentally derived disease threatening numerous residents of rural farming villages along the Danube River in countries of the Balkan Peninsula. This perspective updates recent findings on the identification of AAs in food as a result of the root uptake of free AAs released from the decayed seeds of Aristolochia clematitis L., in combination with their presence and fate in the environment. The potential link between AAs and the high prevalence of chronic kidney diseases in China is also discussed.
Collapse
|
11
|
Chen J, You H, Li Y, Xu Y, He Q, Harris RC. EGF Receptor-Dependent YAP Activation Is Important for Renal Recovery from AKI. J Am Soc Nephrol 2018; 29:2372-2385. [PMID: 30072422 PMCID: PMC6115662 DOI: 10.1681/asn.2017121272] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 06/22/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Increasing evidence indicates that renal recovery from AKI stems from dedifferentiation and proliferation of surviving tubule epithelial cells. Both EGF receptor (EGFR) and the Hippo signaling pathway are implicated in cell proliferation and differentiation, and previous studies showed that activation of EGFR in renal proximal tubule epithelial cells (RPTCs) plays a critical role in recovery from ischemia-reperfusion injury (IRI). In this study, we explored RPTC activation of Yes-associated protein (YAP) and transcriptional coactivator with PDZ binding motif (TAZ), two key downstream effectors of the Hippo pathway, and their potential involvement in recovery from AKI. METHODS We used immunofluorescence to examine YAP expression in kidney biopsy samples from patients with clinical AKI and controls (patients with minimal change disease). Studies of RPTC activation of YAP and TAZ used cultured human RPTCs that were exposed to hypoxia-reoxygenation as well as knockout mice (with inducible deletions of Yap, Taz, or both occurring specifically in RPTCs) that were subjected to bilateral IRI. RESULTS YAP was activated in RPTCs in kidneys from post-AKI patients and post-IRI mouse kidneys. Inhibition of the interaction of YAP and the TEA domain (TEAD) transcription factor complex by verteporfin or conditional deletion of YAP in RPTCs delayed renal functional and structural recovery from IRI, whereas TAZ deletion had no effect. Activation of the EGFR-PI3K-Akt pathway in response to IRI signaled YAP activation, which promoted cell cycle progression. CONCLUSIONS This study shows that EGFR-PI3K-Akt-dependent YAP activation plays an essential role in mediating epithelial cell regeneration during kidney recovery from AKI.
Collapse
Affiliation(s)
- Jianchun Chen
- Department of Veterans Affairs, Nashville, Tennessee; Departments of
- Medicine and
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Huaizhou You
- Medicine and
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China; and
| | - Yan Li
- Medicine and
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | - Raymond C Harris
- Department of Veterans Affairs, Nashville, Tennessee; Departments of
- Medicine and
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee
- Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
12
|
Datta R, Wong A, Camarata T, Tamanna F, Ilahi I, Vasilyev A. Precise Cellular Ablation Approach for Modeling Acute Kidney Injury in Developing Zebrafish. J Vis Exp 2017. [PMID: 28605371 DOI: 10.3791/55606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Acute Kidney Injury (AKI) is a common medical condition with a high mortality rate. With the repair abilities of the kidney, it is possible to restore adequate kidney function after supportive treatment. However, a better understanding of how nephron cell death and repair occur on the cellular level is required to minimize cell death and to enhance the regenerative process. The zebrafish pronephros is a good model system to accomplish this goal because it contains anatomical segments that are similar to the mammalian nephron. Previously, the most common model used to study kidney injury in fish was the pharmacological gentamicin model. However, this model does not allow for precise spatiotemporal control of injury, and hence it is difficult to study cellular and molecular processes involved in kidney repair. To overcome this limitation, this work presents a method through which, in contrast to the gentamicin approach, a specific Green Fuorescent Protein (GFP)-expressing nephron segment can be photoablated using a violet laser light (405 nm). This novel model of AKI provides many advantages that other methods of epithelial injury lack. Its main advantages are the ability to "dial" the level of injury and the precise spatiotemporal control in the robust in vivo animal model. This new method has the potential to significantly advance the level of understanding of kidney injury and repair mechanisms.
Collapse
Affiliation(s)
| | - Ada Wong
- Department of Biomedical Sciences, NYITCOM
| | | | | | | | | |
Collapse
|
13
|
Aquaporin-3 deletion in mice results in renal collecting duct abnormalities and worsens ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1231-1241. [PMID: 28344130 DOI: 10.1016/j.bbadis.2017.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/15/2017] [Accepted: 03/22/2017] [Indexed: 12/15/2022]
Abstract
Aquaporin-3 (AQP3), a transporter of water, glycerol and H2O2, is expressed in basolateral membranes of principal cells in kidney collecting duct. Here, we report that AQP3 deletion in mice affects renal function and modulates renal injury. We found collecting duct hyperplasia and cell swelling in kidneys of adult AQP3 null mice. After mild renal ischemia-reperfusion (IR), AQP3 null mice had significantly greater blood urea nitrogen (57mg/dl) and creatinine (136μM) than wild-type mice (35mg/dl and 48μM, respectively), and showed renal morphological changes, including tubular dilatation, erythrocyte diapedesis and collecting duct incompletion. MPO, MDA and SOD following IR in AQP3 null mice were significantly different from that in wild-type mice (1.7U/g vs 0.8U/g, 3.9μM/g vs 2.4μM/g, 6.4U/mg vs 11U/mg, respectively). Following IR, AQP3 deletion inhibited activation of mitogen-activated protein kinase (MAPK) signaling and produced an increase in the ratios of Bax/Bcl-2, cleaved caspase-3/caspase-3 and p-p53/p53. Studies in transfected MDCK cells showed that AQP3 expression attenuated reduced cell viability following hypoxia-reoxygenation, with reduced apoptosis and increased MAPK signaling. Our results support a novel role for AQP3 in modulating renal injury and suggest the mechanisms involved in protection against hypoxic injury.
Collapse
|
14
|
Stem cell therapy: An emerging modality in glomerular diseases. Cytotherapy 2017; 19:333-348. [PMID: 28089754 DOI: 10.1016/j.jcyt.2016.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/17/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022]
Abstract
The kidney has been considered a highly terminally differentiated organ with low proliferative potential and thus unlikely to undergo regeneration. Glomerular disease progresses to end-stage renal disease (ESRD), which requires dialysis or renal transplantation for better quality of life for patients with ESRD. Because of the shortage of implantable kidneys and complications such as immune rejection, septicemia and toxicity of immunosuppression, kidney transplantation remains a challenge. Therapeutic options available for glomerular disease include symptomatic treatment and strategies to delay progression. In an attempt to develop innovative treatments by promoting the limited capability of regeneration and repair after kidney injury and overcome the progressive pathological process that is uncontrolled with conventional treatment modalities, stem cell-based therapy has emerged as novel intervention due to its ability to inhibit inflammation and promote regeneration. Recent developments in cell therapy have demonstrated promising therapeutic outcomes in terms of restoration of renal structure and function. This review focuses on stem cell therapy approaches for the treatment of glomerular disease, including the various cell sources used and recent advances in preclinical and clinical studies.
Collapse
|
15
|
Wang X, Nichols L, Grunz-Borgmann EA, Sun Z, Meininger GA, Domeier TL, Baines CP, Parrish AR. Fascin2 regulates cisplatin-induced apoptosis in NRK-52E cells. Toxicol Lett 2016; 266:56-64. [PMID: 27989596 DOI: 10.1016/j.toxlet.2016.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/01/2016] [Accepted: 11/27/2016] [Indexed: 01/09/2023]
Abstract
Previous studies have shown that the aging kidney has a marked loss of α(E)-catenin in proximal tubular epithelium. α-Catenin, a key regulator of the actin cytoskeleton, interacts with a variety of actin-binding proteins. Cisplatin-induced loss of fascin2, an actin bundling protein, was observed in cells with a stable knockdown of α(E)-catenin (C2 cells), as well as in aging (24 mon), but not young (4 mon), kidney. Fascin2 co-localized with α-catenin and the actin cytoskeleton in NRK-52E cells. Knockdown of fascin2 increased the susceptibility of tubular epithelial cells to cisplatin-induced injury. Overexpression of fascin2 in C2 cells restored actin stress fibers and attenuated the increased sensitivity of C2 cells to cisplatin-induced apoptosis. Interestingly, fascin2 overexpression attenuated cisplatin-induced mitochondrial dysfunction and oxidative stress in C2 cells. These data demonstrate that fascin2, a putative target of α(E)-catenin, may play important role in preventing cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - LaNita Nichols
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Elizabeth A Grunz-Borgmann
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Christopher P Baines
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Alan R Parrish
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States.
| |
Collapse
|
16
|
Bandodkar AJ, López CS, Vinu Mohan AM, Yin L, Kumar R, Wang J. All-printed magnetically self-healing electrochemical devices. SCIENCE ADVANCES 2016; 2:e1601465. [PMID: 27847875 PMCID: PMC5099985 DOI: 10.1126/sciadv.1601465] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/29/2016] [Indexed: 05/18/2023]
Abstract
The present work demonstrates the synthesis and application of permanent magnetic Nd2Fe14B microparticle (NMP)-loaded graphitic inks for realizing rapidly self-healing inexpensive printed electrochemical devices. The incorporation of NMPs into the printable ink imparts impressive self-healing ability to the printed conducting trace, with rapid (~50 ms) recovery of repeated large (3 mm) damages at the same or different locations without any user intervention or external trigger. The permanent and surrounding-insensitive magnetic properties of the NMPs thus result in long-lasting ability to repair extreme levels of damage, independent of ambient conditions. This remarkable self-healing capability has not been reported for existing man-made self-healing systems and offers distinct advantages over common capsule and intrinsically self-healing systems. The printed system has been characterized by leveraging crystallographic, magnetic hysteresis, microscopic imaging, electrical conductivity, and electrochemical techniques. The real-life applicability of the new self-healing concept is demonstrated for the autonomous repair of all-printed batteries, electrochemical sensors, and wearable textile-based electrical circuits, indicating considerable promise for widespread practical applications and long-lasting printed electronic devices.
Collapse
|
17
|
Thukral SK, Nordone PJ, Hu R, Sullivan L, Galambos E, Fitzpatrick VD, Healy L, Bass MB, Cosenza ME, Afshari CA. Prediction of Nephrotoxicant Action and Identification of Candidate Toxicity-Related Biomarkers. Toxicol Pathol 2016; 33:343-55. [PMID: 15805072 DOI: 10.1080/01926230590927230] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A vast majority of pharmacological compounds and their metabolites are excreted via the urine, and within the complex structure of the kidney, the proximal tubules are a main target site of nephrotoxic compounds. We used the model nephrotoxicants mercuric chloride, 2-bromoethylamine hydrobromide, hexachlorobutadiene, mitomycin, amphotericin, and puromycin to elucidate time- and dose-dependent global gene expression changes associated with proximal tubular toxicity. Male Sprague–Dawley rats were dosed via intraperitoneal injection once daily for mercuric chloride and amphotericin (up to 7 doses), while a single dose was given for all other compounds. Animals were exposed to 2 different doses of these compounds and kidney tissues were collected on day 1, 3, and 7 postdosing. Gene expression profiles were generated from kidney RNA using 17K rat cDNA dual dye microarray and analyzed in conjunction with histopathology. Analysis of gene expression profiles showed that the profiles clustered based on similarities in the severity and type of pathology of individual animals. Further, the expression changes were indicative of tubular toxicity showing hallmarks of tubular degeneration/regeneration and necrosis. Use of gene expression data in predicting the type of nephrotoxicity was then tested with a support vector machine (SVM)-based approach. A SVM prediction module was trained using 120 profiles of total profiles divided into four classes based on the severity of pathology and clustering. Although mitomycin C and amphotericin B treatments did not cause toxicity, their expression profiles were included in the SVM prediction module to increase the sample size. Using this classifier, the SVM predicted the type of pathology of 28 test profiles with 100% selectivity and 82% sensitivity. These data indicate that valid predictions could be made based on gene expression changes from a small set of expression profiles. A set of potential biomarkers showing a time- and dose-response with respect to the progression of proximal tubular toxicity were identified. These include several transporters ( Slc21a2, Slc15, Slc34a2), Kim 1, IGFbp-1, osteopontin, α -fibrinogen, and Gstα.
Collapse
|
18
|
Iwakura T, Fujigaki Y, Fujikura T, Ohashi N, Kato A, Yasuda H. Acquired resistance to rechallenge injury after acute kidney injury in rats is associated with cell cycle arrest in proximal tubule cells. Am J Physiol Renal Physiol 2016; 310:F872-84. [PMID: 26823281 DOI: 10.1152/ajprenal.00380.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 01/27/2016] [Indexed: 01/26/2023] Open
Abstract
Rats that have recovered from severe proximal tubule (PT) injury induced by uranyl acetate (UA), a toxic stimulus, developed resistance to subsequent UA treatment. We investigated cell cycle status and progression in PT cells in relation to this acquired resistance. Fourteen days after pretreatment with saline (vehicle group) or UA [acute kidney injury (AKI) group], rats were injected with UA or lead acetate (a proliferative stimulus). Cell cycle status (G0/G1/S/G2/M) was analyzed by flow cytometry. The expression of cell cycle markers, cyclin-dependent kinase inhibitors, and phenotypic markers were examined by immunohistochemistry. Cell cycle status in PT cells in the AKI group was comparable to those of the vehicle group. However, more early G1-phase cells (cyclin D1- or Ki67-) and p21+ or p27+ cells were found in the PT of the AKI group than in that of the vehicle group. UA induced G1 arrest and inhibited S phase progression with earlier dedifferentiation and less apoptosis in PT cells of the AKI group. Lead acetate induced proliferation without dedifferentiation but with delayed G0-G1 transition and inhibited S phase progression in PT cells in the AKI group. Sustained p21 and increased p27 expression in PT cells were found in the AKI group in response to UA and lead acetate. PT cells in the AKI group inhibited cell cycle progression by enhanced G1 arrest, probably via p21/p27 modulation as an injury or proliferation response, resulting in cytoresistance to rechallenge injury.
Collapse
Affiliation(s)
- Takamasa Iwakura
- Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshihide Fujigaki
- Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Tomoyuki Fujikura
- Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naro Ohashi
- Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akihiko Kato
- Blood Purification Unit, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideo Yasuda
- Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
19
|
Ischemic postconditioning inhibits apoptosis of renal cells following reperfusion: a novel in vitro model. Int Urol Nephrol 2015; 47:1067-74. [DOI: 10.1007/s11255-015-0997-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/21/2015] [Indexed: 11/26/2022]
|
20
|
Lawson J, Elliott J, Wheeler-Jones C, Syme H, Jepson R. Renal fibrosis in feline chronic kidney disease: known mediators and mechanisms of injury. Vet J 2014; 203:18-26. [PMID: 25475166 DOI: 10.1016/j.tvjl.2014.10.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/10/2014] [Accepted: 10/11/2014] [Indexed: 01/13/2023]
Abstract
Chronic kidney disease (CKD) is a common medical condition of ageing cats. In most cases the underlying aetiology is unknown, but the most frequently reported pathological diagnosis is renal tubulointerstitial fibrosis. Renal fibrosis, characterised by extensive accumulation of extra-cellular matrix within the interstitium, is thought to be the final common pathway for all kidney diseases and is the pathological lesion best correlated with function in both humans and cats. As a convergent pathway, renal fibrosis provides an ideal target for the treatment of CKD and knowledge of the underlying fibrotic process is essential for the future development of novel therapies. There are many mediators and mechanisms of renal fibrosis reported in the literature, of which only a few have been investigated in the cat. This article reviews the process of renal fibrosis and discusses the most commonly cited mediators and mechanisms of progressive renal injury, with particular focus on the potential significance to feline CKD.
Collapse
Affiliation(s)
- Jack Lawson
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| | - Jonathan Elliott
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Caroline Wheeler-Jones
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Harriet Syme
- Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - Rosanne Jepson
- Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| |
Collapse
|
21
|
McCampbell KK, Springer KN, Wingert RA. Analysis of nephron composition and function in the adult zebrafish kidney. J Vis Exp 2014:e51644. [PMID: 25145398 PMCID: PMC4459603 DOI: 10.3791/51644] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The zebrafish model has emerged as a relevant system to study kidney development, regeneration and disease. Both the embryonic and adult zebrafish kidneys are composed of functional units known as nephrons, which are highly conserved with other vertebrates, including mammals. Research in zebrafish has recently demonstrated that two distinctive phenomena transpire after adult nephrons incur damage: first, there is robust regeneration within existing nephrons that replaces the destroyed tubule epithelial cells; second, entirely new nephrons are produced from renal progenitors in a process known as neonephrogenesis. In contrast, humans and other mammals seem to have only a limited ability for nephron epithelial regeneration. To date, the mechanisms responsible for these kidney regeneration phenomena remain poorly understood. Since adult zebrafish kidneys undergo both nephron epithelial regeneration and neonephrogenesis, they provide an outstanding experimental paradigm to study these events. Further, there is a wide range of genetic and pharmacological tools available in the zebrafish model that can be used to delineate the cellular and molecular mechanisms that regulate renal regeneration. One essential aspect of such research is the evaluation of nephron structure and function. This protocol describes a set of labeling techniques that can be used to gauge renal composition and test nephron functionality in the adult zebrafish kidney. Thus, these methods are widely applicable to the future phenotypic characterization of adult zebrafish kidney injury paradigms, which include but are not limited to, nephrotoxicant exposure regimes or genetic methods of targeted cell death such as the nitroreductase mediated cell ablation technique. Further, these methods could be used to study genetic perturbations in adult kidney formation and could also be applied to assess renal status during chronic disease modeling.
Collapse
|
22
|
Palmyre A, Lee J, Ryklin G, Camarata T, Selig MK, Duchemin AL, Nowak P, Arnaout MA, Drummond IA, Vasilyev A. Collective epithelial migration drives kidney repair after acute injury. PLoS One 2014; 9:e101304. [PMID: 25010471 PMCID: PMC4092191 DOI: 10.1371/journal.pone.0101304] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 06/04/2014] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) is a common and significant medical problem. Despite the kidney’s remarkable regenerative capacity, the mortality rate for the AKI patients is high. Thus, there remains a need to better understand the cellular mechanisms of nephron repair in order to develop new strategies that would enhance the intrinsic ability of kidney tissue to regenerate. Here, using a novel, laser ablation-based, zebrafish model of AKI, we show that collective migration of kidney epithelial cells is a primary early response to acute injury. We also show that cell proliferation is a late response of regenerating kidney epithelia that follows cell migration during kidney repair. We propose a computational model that predicts this temporal relationship and suggests that cell stretch is a mechanical link between migration and proliferation, and present experimental evidence in support of this hypothesis. Overall, this study advances our understanding of kidney repair mechanisms by highlighting a primary role for collective cell migration, laying a foundation for new approaches to treatment of AKI.
Collapse
Affiliation(s)
- Aurélien Palmyre
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Jeongeun Lee
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Biomedical Sciences, NYIT COM, Old Westbury, New York, United States of America
| | - Gennadiy Ryklin
- Department of Biomedical Sciences, NYIT COM, Old Westbury, New York, United States of America
| | - Troy Camarata
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Martin K. Selig
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Anne-Laure Duchemin
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Paul Nowak
- Department of Biomedical Sciences, NYIT COM, Old Westbury, New York, United States of America
| | - M. Amin Arnaout
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Developmental and Regenerative Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Iain A. Drummond
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aleksandr Vasilyev
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Biomedical Sciences, NYIT COM, Old Westbury, New York, United States of America
- * E-mail:
| |
Collapse
|
23
|
Nichols LA, Grunz-Borgmann EA, Wang X, Parrish AR. A role for the age-dependent loss of α(E)-catenin in regulation of N-cadherin expression and cell migration. Physiol Rep 2014; 2:2/6/e12039. [PMID: 24920123 PMCID: PMC4208646 DOI: 10.14814/phy2.12039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The aging kidney has a decreased ability to repair following acute kidney injury. Previous studies from our laboratory have demonstrated a loss in α‐catenin expression in the aging rat kidney. We hypothesize that loss of α‐catenin expression in tubular epithelial cells may induce changes that result in a decreased repair capacity. In these studies, we demonstrate that decreased α‐catenin protein expression is detectable as early as 20 months of age in male Fischer 344 rats. Protein loss is also observed in aged nonhuman primate kidneys, suggesting that this is not a species‐specific response. In an effort to elucidate alterations due to the loss of α‐catenin, we generated NRK‐52E cell lines with stable knockdown of α(E)‐catenin (C2 cells). Interestingly, C2 cells had decreased expression of N‐cadherin, decreased cell–cell adhesion, and increased monolayer permeability. C2 had deficits in wound repair, due to alterations in cell migration. Analysis of gene expression in the migrating control cells indicated that expression of N‐cadherin and N‐CAM was increased during repair. In migrating C2 cells, expression of N‐CAM was also increased, but the expression of N‐cadherin was not upregulated. Importantly, a blocking antibody against N‐cadherin inhibited repair in NRK‐52E cells, suggesting an important role in repair. Taken together, these data suggest that loss of α‐catenin, and the subsequent downregulation of N‐cadherin expression, is a mechanism underlying the decreased migration of tubular epithelial cells that contributes to the inability of the aging kidney to repair following injury. Aging is associated with loss of α‐catenin and N‐cadherin expression in the kidney. In these studies, we demonstrate that α‐catenin regulates, in part, N‐cadherin expression and migration in tubular epithelial cells.
Collapse
Affiliation(s)
- LaNita A Nichols
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| | | | - Xinhui Wang
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Alan R Parrish
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
24
|
Bakker PJ, Butter LM, Claessen N, Teske GJD, Sutterwala FS, Florquin S, Leemans JC. A tissue-specific role for Nlrp3 in tubular epithelial repair after renal ischemia/reperfusion. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2013-22. [PMID: 24823805 DOI: 10.1016/j.ajpath.2014.04.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/25/2014] [Accepted: 04/08/2014] [Indexed: 10/25/2022]
Abstract
Ischemia/reperfusion injury is a major cause of acute kidney injury. Improving renal repair would represent a therapeutic strategy to prevent renal dysfunction. The innate immune receptor Nlrp3 is involved in tissue injury, inflammation, and fibrosis; however, its role in repair after ischemia/reperfusion is unknown. We address the role of Nlrp3 in the repair phase of renal ischemia/reperfusion and investigate the relative contribution of leukocyte- versus renal-associated Nlrp3 by studying bone marrow chimeric mice. We found that Nlrp3 expression was most profound during the repair phase. Although Nlrp3 expression was primarily expressed by leukocytes, both leukocyte- and renal-associated Nlrp3 was detrimental to renal function after ischemia/reperfusion. The Nlrp3-dependent cytokine IL-1β remained unchanged in kidneys of all mice. Leukocyte-associated Nlrp3 negatively affected tubular apoptosis in mice that lacked Nlrp3 expression on leukocytes, which correlated with reduced macrophage influx. Nlrp3-deficient (Nlrp3KO) mice with wild-type bone marrow showed an improved repair response, as seen by a profound increase in proliferating tubular epithelium, which coincided with increased hepatocyte growth factor expression. In addition, Nlrp3KO tubular epithelial cells had an increased repair response in vitro, as seen by an increased ability of an epithelial monolayer to restore its structural integrity. In conclusion, Nlrp3 shows a tissue-specific role in which leukocyte-associated Nlrp3 is associated with tubular apoptosis, whereas renal-associated Nlrp3 impaired wound healing.
Collapse
Affiliation(s)
- Pieter J Bakker
- Department of Pathology, Academic Medical Center, Amsterdam, the Netherlands.
| | - Loes M Butter
- Department of Pathology, Academic Medical Center, Amsterdam, the Netherlands
| | - Nike Claessen
- Department of Pathology, Academic Medical Center, Amsterdam, the Netherlands
| | | | | | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, Amsterdam, the Netherlands; Department of Pathology, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Jaklien C Leemans
- Department of Pathology, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Herrera M, Mirotsou M. Stem cells: potential and challenges for kidney repair. Am J Physiol Renal Physiol 2013; 306:F12-23. [PMID: 24197069 DOI: 10.1152/ajprenal.00238.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal damage resulting from acute and chronic kidney injury poses an important problem to public health. Currently, patients with end-stage renal disease rely solely on kidney transplantation or dialysis for survival. Emerging therapies aiming to prevent and reverse kidney damage are thus in urgent need. Although the kidney was initially thought to lack the capacity for self-repair, several studies have indicated that this might not be the case; progenitor and stem cells appear to play important roles in kidney repair under various pathological conditions. In this review, we summarize recent findings on the role of progenitor/stem cells on kidney repair as well as discuss their potential as a therapeutic approach for kidney diseases.
Collapse
Affiliation(s)
- Marcela Herrera
- Division of Cardiology, Genome Research Bldg. II, Rm. 4022, 210 Research Drive, Duke Univ. Medical Center, Durham, NC 27710.
| | | |
Collapse
|
26
|
Ranganathan P, Jayakumar C, Navankasattusas S, Li DY, Kim IM, Ramesh G. UNC5B receptor deletion exacerbates tissue injury in response to AKI. J Am Soc Nephrol 2013; 25:239-49. [PMID: 24115477 DOI: 10.1681/asn.2013040418] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Netrin-1 regulates cell survival and apoptosis by activation of its receptors, including UNC5B. However, the in vivo role of UNC5B in cell survival during cellular stress and tissue injury is unknown. We investigated the role of UNC5B in cell survival in response to stress using mice heterozygously expressing the UNC5B gene (UNC5B(-/flox)) and mice with targeted homozygous deletion of UNC5B in kidney epithelial cells (UNC5B(-/flox/GGT-cre)). Mice were subjected to two different models of organ injury: ischemia reperfusion injury of the kidney and cisplatin-induced nephrotoxicity. Both mouse models of UNC5B depletion had normal organ function and histology under basal conditions. After AKI, however, UNC5B(-/flox/GGT-cre) mice exhibited significantly worse renal function and damage, increased tubular apoptosis, enhanced p53 activation, and exacerbated inflammation compared with UNC5B(-/flox) and wild-type mice. shRNA-mediated suppression of UNC5B expression in cultured tubular epithelial cells exacerbated cisplatin-induced cell death in a p53-dependent manner and blunted Akt phosphorylation. Inhibition of PI3 kinase similarly exacerbated cisplatin-induced apoptosis; in contrast, overexpression of UNC5B reduced cisplatin-induced apoptosis in these cells. Taken together, these results show that the netrin-1 receptor UNC5B plays a critical role in cell survival and kidney injury through Akt-mediated inactivation of p53 in response to stress.
Collapse
Affiliation(s)
- Punithavathi Ranganathan
- Department of Medicine and Vascular Biology Center, Georgia Regents University, Augusta, Georgia; and
| | | | | | | | | | | |
Collapse
|
27
|
Rostami Z, Einollahi B, Ghadiani MH. Does living donor hyperoxia have an impact on kidney graft function after transplantation? Nephrourol Mon 2013; 5:835-9. [PMID: 24282796 PMCID: PMC3830912 DOI: 10.5812/numonthly.11870] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 05/06/2013] [Indexed: 11/16/2022] Open
Abstract
Background Improvement in the outcome of organ transplantation is related to advances in patient selection criteria, organ preservation, operative techniques, perioperative care and efficacy of immunosuppressive agents. Objectives We aimed to evaluate the effects of higher levels of arterial PaO2 in donors on DGF (delayed graft function). Patients and Methods Forty patients over 18 years old with stage 4-5 chronic kidney disease (CKD) who received a kidney from living donors were enrolled. They were randomly grouped in to the case (n = 17) and control (n = 23) groups and were followed for 2 weeks after transplantation. Donors were exposed to 60% oxygen for at least 2 hours with a face-mask (venture mask) for 2 consecutive days before transplantation until arterial oxygen pressure increased in arterial blood gas to 200 mmHg. Neutrophil gelatinase associated lipocalin (NGAL), Interleuk-18 (IL-18), tumor necrosis factor- α (TNF-α) and transforming growth factor–β (TGF-β) could be good biomarkers for early diagnosis of kidney injury in renal transplant recipients; we assessed kidney function with these biomarkers. Results Forty living kidney transplantations including 17 cases and 23 controls were performed; female gender was more prevalent in recipients (n = 16, 40%). The mean age of recipients was 36.1 ± 12.4 (18-67) years old. DGF was detected in 2 (5.95%) individuals, from whom one was in the case group and the other one in the control group. In the univariate analysis, there was no significant correlation between age and biomarkers in urine and serum unless for the second serum NGAL (P = 0.02, r = -0.06) and second urine IL 18 (P = 0.03, r = -0.5) which had a negative correlation, and first urine TNF α (P = 0.02, r = 0.7) which had a positive correlation. Conclusions Oxygen therapy in the case group had no significant impact on protection from DGF.
Collapse
Affiliation(s)
- Zohreh Rostami
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Zohreh Rostami, Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran. Tel: +98-9121544897, Fax: +98-2181262073, E-mail:
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | | |
Collapse
|
28
|
Choi YH, McNally BT, Igarashi P. Zyxin regulates migration of renal epithelial cells through activation of hepatocyte nuclear factor-1β. Am J Physiol Renal Physiol 2013; 305:F100-10. [PMID: 23657850 DOI: 10.1152/ajprenal.00582.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is an epithelial tissue-specific transcription factor that regulates gene expression in the kidney, liver, pancreas, intestine, and other organs. Mutations of HNF-1β in humans produce renal cysts and congenital kidney anomalies. Here, we identify the LIM-domain protein zyxin as a novel binding partner of HNF-1β in renal epithelial cells. Zyxin shuttles to the nucleus where it colocalizes with HNF-1β. Immunoprecipitation of zyxin in leptomycin B-treated cells results in coprecipitation of HNF-1β. The protein interaction requires the second LIM domain of zyxin and two distinct domains of HNF-1β. Overexpression of zyxin stimulates the transcriptional activity of HNF-1β, whereas small interfering RNA silencing of zyxin inhibits HNF-1β-dependent transcription. Epidermal growth factor (EGF) induces translocation of zyxin into the nucleus and stimulates HNF-1β-dependent promoter activity. The EGF-mediated nuclear translocation of zyxin requires activation of Akt. Expression of dominant-negative mutant HNF-1β, knockdown of zyxin, or inhibition of Akt inhibits EGF-stimulated cell migration. These findings reveal a novel pathway by which extracellular signals are transmitted to the nucleus to regulate the activity of a transcription factor that is essential for renal epithelial differentiation.
Collapse
Affiliation(s)
- Yun-Hee Choi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | |
Collapse
|
29
|
Kidney. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
30
|
Wen D, Ni L, You L, Zhang L, Gu Y, Hao CM, Chen J. Upregulation of nestin in proximal tubules may participate in cell migration during renal repair. Am J Physiol Renal Physiol 2012; 303:F1534-44. [PMID: 22993065 DOI: 10.1152/ajprenal.00083.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The characteristics of renal tubular progenitor/precursor cells and the role of renal tubule regeneration in the repair of remnant kidneys (RKs) after nephrectomy are not well known. In the present study of a murine model of subtotal nephrectomy, we used immunofluorescence (IF), immunoblot analysis, and in situ hybridization methods to demonstrate that nestin expression was transiently upregulated in tubule cells near the incision edges of RKs. The nestin-positive tubules were immature proximal tubules that colabeled with lotus tetragonolobus agglutinin but not with markers of mature tubules (aquaporin-1, Tamm-Horsfall protein, and aquaporin-2). In addition, many of the nestin-expressing tubule cells were actively proliferative cells, as indicated by colabeling with bromodeoxyuridine. Double-label IF and immunoblot analysis also showed that the upregulation of tubular nestin was associated with enhanced transforming growth factor-β1 (TGF-β1) expression in the incision edge of RKs but not α-smooth muscle actin, which is a marker of fibrosis. In cultured human kidney proximal tubule cells (HKC), immunoblot analysis indicated that TGF-β1 induced nestin expression and loss of E-cadherin expression, suggesting an association of nestin expression and cellular dedifferentiation. Knockdown of nestin expression by a short hairpin RNA-containing plasmid led to decreased migration of HKC cells that were induced by TGF-β1. Taken together, our results suggest that the tubule repair that occurs during the recovery process following nephrectomy may involve TGF-β1-induced nestin expression in immature renal proximal tubule cells and the promotion of renal cell migration.
Collapse
Affiliation(s)
- Donghai Wen
- Division of Nephrology, Huashan Hospital, Shanghai Medical College, Fudan Univ., Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The kidney is widely regarded as an organ without regenerative abilities. However, in recent years this dogma has been challenged on the basis of observations of kidney recovery following acute injury, and the identification of renal populations that demonstrate stem cell characteristics in various species. It is currently speculated that the human kidney can regenerate in some contexts, but the mechanisms of renal regeneration remain poorly understood. Numerous controversies surround the potency, behaviour and origins of the cell types that are proposed to perform kidney regeneration. The present review explores the current understanding of renal stem cells and kidney regeneration events, and examines the future challenges in using these insights to create new clinical treatments for kidney disease.
Collapse
|
32
|
Protective effect of Urtica dioica L. on renal ischemia/reperfusion injury in rat. J Mol Histol 2012; 43:691-8. [DOI: 10.1007/s10735-012-9436-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 06/21/2012] [Indexed: 10/28/2022]
|
33
|
Chen J, Chen JK, Harris RC. Deletion of the epidermal growth factor receptor in renal proximal tubule epithelial cells delays recovery from acute kidney injury. Kidney Int 2012; 82:45-52. [PMID: 22418982 PMCID: PMC3376190 DOI: 10.1038/ki.2012.43] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/27/2011] [Accepted: 12/20/2011] [Indexed: 12/13/2022]
Abstract
To determine the role of epidermal growth factor receptor (EGFR) activation in renal functional and structural recovery from acute kidney injury (AKI), we generated mice with a specific EGFR deletion in the renal proximal tubule (EGFR(ptKO)). Ischemia-reperfusion injury markedly activated EGFR in control littermate mice; however, this was inhibited in either the knockout or wild-type mice given erlotinib, a specific EGFR tyrosine kinase inhibitor. Blood urea nitrogen and serum creatinine increased to a comparable level in EGFR(ptKO) and control mice 24 h after reperfusion, but the subsequent rate of renal function recovery was markedly slowed in the knockout mice. Twenty-four hours after reperfusion, both the knockout and the inhibitor-treated mice had a similar degree of histologic renal injury as control mice, but at day 6 there was minimal evidence of injury in the control mice while both EGFR(ptKO) and erlotinib-treated mice still had persistent proximal tubule dilation, epithelial simplification, and cast formation. Additionally, renal cell proliferation was delayed due to decreased ERK and Akt signaling. Thus, our studies provide both genetic and pharmacologic evidence that proximal tubule EGFR activation plays an important role in the recovery phase after acute kidney injury.
Collapse
Affiliation(s)
- Jianchun Chen
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.
| | | | | |
Collapse
|
34
|
Ikari A, Takiguchi A, Atomi K, Sato T, Sugatani J. Decrease in claudin-2 expression enhances cell migration in renal epithelial Madin-Darby canine kidney cells. J Cell Physiol 2011; 226:1471-8. [PMID: 20717932 DOI: 10.1002/jcp.22386] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Migration of renal epithelial cells increases after renal tubular damage, but its mechanism has not been clarified in detail. Hyperosmotic stress increased a cellular injury concomitant with a decrease in mRNA and protein expression of claudin-2 in renal tubular epithelial Madin-Darby canine kidney cells. We hypothesized that claudin-2 is involved in the regulation of cell migration. To knockdown claudin-2 expression, we made the cells expressing doxycycline-inducible claudin-2 shRNA vector. Claudin-2 knockdown affected neither the endogenous expression levels of claudin-1, -3, -4, and -7 nor the Triton X-100 solubility of these claudins. Transepithelial electrical resistance was increased by claudin-2 knockdown without affecting permeability to FITC-dextran (4,000 Da). BrdU incorporation assay and cell counting revealed that cell proliferation and viability are unaffected by claudin-2 knockdown. In the wound-healing assay, the recovery rate of wound area was increased by claudin-2 knockdown. The mRNA expression and activity of matrix metalloproteinase-9 (MMP-9) were increased by claudin-2 knockdown. A selective MMP-9 inhibitor suppressed cell migration in the claudin-2 knockdown cells. Hyperosmotic stress increased the expression and activity of MMP-9, which were inhibited by claudin-2 overexpression. These results suggest that the decrease in claudin-2 expression enhances cell migration mediated by the increase in the expression and activity of MMP-9.
Collapse
Affiliation(s)
- Akira Ikari
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| | | | | | | | | |
Collapse
|
35
|
Kidney. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
36
|
Gu JW, Manning RD, Young E, Shparago M, Sartin B, Bailey AP. Vascular endothelial growth factor receptor inhibitor enhances dietary salt-induced hypertension in Sprague-Dawley rats. Am J Physiol Regul Integr Comp Physiol 2009; 297:R142-8. [PMID: 19420288 DOI: 10.1152/ajpregu.90972.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Clinical evidence links the inhibition of VEGF to hypertension. However, the mechanisms by which VEGF affects the pathogenesis of hypertension remain in question. We determined 1) whether administration of VEGF receptor inhibitor SU5416 enhances dietary salt-induced hypertension in Sprague-Dawley (SD) rats, and 2) whether VEGF or SU5416 directly affects proliferation of cultured human renal proximal tubular epithelial cells (HRPTEC) and endothelial nitric oxide synthase (eNOS) expression in cultured human glomerular microvessel endothelial cells (HGMEC). Ten 10-wk-old male SD rats received a high sodium diet (HS; 8%) and the other 10 SD rats received a normal sodium diet (NS; 0.5%) for 4 wks. After 2 wks of the dietary program, five rats were administered with SU5416 at 10 mg x kg(-1) x day(-1) ip or DMSO (vehicle) for 14 days in HS and NS groups. Mean arterial pressure was significantly higher in rats treated with SU5416, as opposed to those treated with DMSO and fed with HS for 4 wk (157.6 +/- 3.9 vs. 125.9 +/- 4.3 mmHg, P < 0.01). Increased proteinuria and albuminuria were associated with marked renal histological abnormalities in HS group with SU5416 administration, compared with those in the vehicle HS group. 3H-thymidine incorporation assay showed that SU5416 blocked the actions of both exogenous and endogenous VEGF on the proliferation of HRPTEC. VEGF (10 ng/ml) significantly increased eNOS protein levels by 29% in cultured HGMEC, but its action was completely abolished by SU5416. These results suggest that VEGF receptor inhibition enhances dietary salt-induced hypertension and kidney injury, possibly by direct damage on renal cells and decreasing NO production by eNOS.
Collapse
Affiliation(s)
- Jian-Wei Gu
- Dept. of Physiology and Biophysics, Univ. of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216-4505, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Fujigaki Y, Sakakima M, Sun Y, Fujikura T, Tsuji T, Yasuda H, Hishida A. Cell division and phenotypic regression of proximal tubular cells in response to uranyl acetate insult in rats. Nephrol Dial Transplant 2009; 24:2686-92. [PMID: 19395729 DOI: 10.1093/ndt/gfp199] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We examined whether dedifferentiation is necessary for cell division of proximal tubule (PT) cells after acute PT injury. METHODS Rats were injected with a low (0.2 mg/kg) or high (4 mg/kg) dose of uranyl acetate (UA) to induce acute PT injury. Proliferating PT cells were labelled with bromodeoxyuridine (BrdU) before sacrifice. Renal tissues were examined by double labelling of BrdU and megalin, aquaporin 1 (AQP1), Na(+)-K(+)ATPase or vimentin, and by immunoelectron microscopy for BrdU+ cells. RESULTS Under normal conditions, BrdU+ PT cells were positive for the PT phenotype (megalin-, AQP1- and Na(+)-K(+)ATPase positive and vimentine negative, a mesenchymal marker). Low-dose UA induced focal PT injury, and BrdU+ initially proliferating PT cells were found in the proximal three quarters of the S3 segment of nephron as early as 12 h, which maintained the PT phenotype and were vimentin negative. Proliferating PT cells showed low expression of the PT cell protein phenotype from Day 2 to Day 5 with vimentin expression from Day 2. High-dose UA induced severe PT injury in the proximal three quarters of the S3 segment by Day 5. BrdU+ initially proliferating PT cells, which were found in distal areas of the S3 segment as early as Day 2, showed low expression of the PT protein phenotype but were vimentin positive. Immunoelectron microscopy showed mature PT morphology for BrdU+ PT cells in control rats. BrdU+ initially proliferating PT cells showed a relatively mature phenotype after low-dose UA in- sult but an immature phenotype after high-dose UA insult. CONCLUSIONS PT cells can initiate cell division without de- differentiation after mild PT injury by low-dose UA insult.
Collapse
Affiliation(s)
- Yoshihide Fujigaki
- First Department of Medicine, Hamamatsu University School of Medicine, Japan.
| | | | | | | | | | | | | |
Collapse
|
38
|
Zhou T, Chou J, Watkins PB, Kaufmann WK. Toxicogenomics: transcription profiling for toxicology assessment. EXS 2009; 99:325-66. [PMID: 19157067 DOI: 10.1007/978-3-7643-8336-7_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Toxicogenomics, the application of transcription profiling to toxicology, has been widely used for elucidating the molecular and cellular actions of chemicals and other environmental stressors on biological systems, predicting toxicity before any functional damages, and classification of known or new toxicants based on signatures of gene expression. The success of a toxicogenomics study depends upon close collaboration among experts in different fields, including a toxicologist or biologist, a bioinformatician, statistician, physician and, sometimes, mathematician. This review is focused on toxicogenomics studies, including transcription profiling technology, experimental design, significant gene extraction, toxicological results interpretation, potential pathway identification, database input and the applications of toxicogenomics in various fields of toxicological study.
Collapse
Affiliation(s)
- Tong Zhou
- Center for Drug Safety Sciences, The Hamner Institutes for Health Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, NC, USA.
| | | | | | | |
Collapse
|
39
|
Israni AK, Li N, Cizman BB, Snyder J, Abrams J, Joffe M, Rebbeck T, Feldman HI. Association of donor inflammation- and apoptosis-related genotypes and delayed allograft function after kidney transplantation. Am J Kidney Dis 2008; 52:331-9. [PMID: 18640487 PMCID: PMC2562522 DOI: 10.1053/j.ajkd.2008.05.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 05/07/2008] [Indexed: 11/11/2022]
Abstract
BACKGROUND Delayed renal allograft survival (delayed graft function [DGF]) after deceased donor kidney transplantation is associated with an increased risk of allograft loss. Inflammatory response and apoptosis are associated with increased risk of DGF. STUDY DESIGN Cross-sectional study. SETTING & PARTICIPANTS We first recruited 616 recipients of kidneys from 512 deceased kidney donors, and donor DNA was genotyped. These recipients, who were included in a prospective cohort study of 9 transplant centers in the Delaware Valley region, had their DGF outcome obtained through medical record abstraction. We then identified the recipient (n = 349) of the contralateral deceased kidney donor, if not part of the cohort, through the US Renal Data System registry. The final cohort consisted of 965 recipients of deceased donor kidneys from 512 donors. PREDICTORS Donor single-nucleotide polymorphisms in genes for tumor necrosis factor alpha (TNF), transforming growth factor beta1 (TGFB1), interleukin 10 (IL10), p53 (TP53), and heme oxygenase 1 (HMOX1). OUTCOMES DGF, defined as the need for dialysis therapy in the first week posttransplantation. Secondary outcomes included acute rejection and estimated glomerular filtration rate. MEASUREMENTS Information for DGF, acute rejection, and estimated glomerular filtration rate for recipients in the Delaware Valley Cohort was obtained through medical record abstraction. For other recipients, information for DGF was obtained from United Network for Organ Sharing forms and Centers for Medicare & Medicaid Services claims in the US Renal Data System registry. RESULTS No association was detected between the TGFB1, IL10, TP53, and HMOX1 genes and DGF. The G allele of the TNF polymorphism rs3093662 was associated with DGF in an adjusted analysis (odds ratio, 1.85 compared with A allele; 95% confidence interval, 1.16 to 2.96; P = 0.01). However, this association did not achieve statistical significance after adjusting for multiple comparisons. LIMITATIONS Inadequate sample size for infrequent genotypes and multiple comparisons. CONCLUSION Because of the low frequency of donor single-nucleotide polymorphisms of interest, a larger sample size and replication are necessary to confirm these findings on the association of donor genotypes with DGF.
Collapse
Affiliation(s)
- Ajay K. Israni
- Department of Medicine, Hennepin County Medical Center, University of Minnesota
- Department of Epidemiology & Community Health, University of Minnesota
| | - Na Li
- Department of Biostatistics, University of Minnesota
| | - Bojana B. Cizman
- Center for Clinical Epidemiology & Biostatistics, University of Pennsylvania
| | - Jon Snyder
- Chronic Disease Research Group, Minneapolis Medical Research Foundation
| | | | - Marshall Joffe
- Center for Clinical Epidemiology & Biostatistics, University of Pennsylvania
| | - Timothy Rebbeck
- Center for Clinical Epidemiology & Biostatistics, University of Pennsylvania
| | - Harold I. Feldman
- Center for Clinical Epidemiology & Biostatistics, University of Pennsylvania
- Department of Medicine, University of Pennsylvania
| |
Collapse
|
40
|
Hallman MA, Zhuang S, Schnellmann RG. Regulation of dedifferentiation and redifferentiation in renal proximal tubular cells by the epidermal growth factor receptor. J Pharmacol Exp Ther 2008; 325:520-8. [PMID: 18270318 DOI: 10.1124/jpet.107.134031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Repair of injured renal epithelium is thought to be mediated by surviving renal proximal tubular cells (RPTC) that must dedifferentiate to allow the proliferation and migration necessary for epithelial regeneration. RPTC then redifferentiate to restore tubular structure and function. Current models suggest that epidermal growth factor receptor (EGFR) activation is required for dedifferentiation characterized by enhanced vimentin expression, decreased N-cadherin expression, spindle morphology, and loss of apical-basal polarity after injury. Because an in vitro model of RPTC redifferentiation has not been reported, and the mechanism(s) of redifferentiation has not been determined, we used rabbit RPTC in primary cultures to address these issues. H2O2 induced the dedifferentiated phenotype that persisted >48 h; redifferentiation occurred spontaneously in the absence of exogenous growth factors after 72 to 120 h. Phosphorylation of two tyrosine residues of EGFR increased 12 to 24 h, peaked at 24 h, and declined to basal levels by 48 h after injury. EGFR inhibition during dedifferentiation restored epithelial morphology and apical-basal polarity, and it decreased vimentin expression to control levels 24 h later. In contrast, exogenous epidermal growth factor addition increased vimentin expression and potentiated spindle morphology. p38 mitogen-activated protein kinase (MAPK) and transforming growth factor (TGF)-beta receptor inhibitors did not affect redifferentiation after H2O2 injury. Similar results were observed in a mechanical injury model. These experiments represent a new model for the investigation of RPTC redifferentiation after acute injury and identify a key regulator of redifferentiation: EGFR, independent of p38 MAPK and the TGF-beta receptor.
Collapse
Affiliation(s)
- Mark A Hallman
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 280 Calhoun St., P.O.B. 250140, Charleston, SC 29425, USA
| | | | | |
Collapse
|
41
|
Rasbach KA, Schnellmann RG. Isoflavones promote mitochondrial biogenesis. J Pharmacol Exp Ther 2008; 325:536-43. [PMID: 18267976 DOI: 10.1124/jpet.107.134882] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial damage is often both the cause and outcome of cell injury resulting from a variety of toxic insults, hypoxia, or trauma. Increasing mitochondrial biogenesis after renal proximal tubular cell (RPTC) injury accelerated the recovery of mitochondrial and cellular functions (Biochem Biophys Res Commun 355:734-739, 2007). However, few pharmacological agents are known to increase mitochondrial biogenesis. We report that daidzein, genistein, biochanin A, formononetin, 3-(2',4'-dichlorophenyl)-7-hydroxy-4H-chromen-4-one (DCHC), 7-hydroxy-4H-chromen-4-one (7-C), 4'7-dimethoxyisoflavone (4',7-D), and 5,7,4'-trimethoxyisoflavone (5,7,4'-T) increased peroxisome proliferator-activated receptor gamma coactivator (PGC)-1alpha expression and resulted in mitochondrial biogenesis as indicated by increased expression of ATP synthase beta and ND6, and 1.5-fold increases in respiration and ATP in RPTC. Inhibition of estrogen receptors with ICI182780 (fulvestrant) had no effect on daidzein-induced mitochondrial biogenesis. The isoflavone derivatives showed differential effects on the activation and expression of sirtuin (SIRT)1, a deacetylase and activator of PGC-1alpha. Daidzein and formononetin induced the expression of SIRT1 in RPTC and the activation of recombinant SIRT1, whereas DCHC and 7-C only induced the activation of recombinant SIRT1. In contrast, genistein, biochanin A, 4',7-D, and 5,7,4'-T only increased SIRT1 expression in RPTC. We have identified a series of substituted isoflavones that produce mitochondrial biogenesis through PGC1alpha and increased SIRT1 activity and/or expression, independently of the estrogen receptor. Furthermore, different structural components are responsible for the activities of isoflavones: the hydroxyl group at position 7 is required SIRT1 activation, a hydroxyl group at position 5 blocks SIRT1 activation, and the loss of the phenyl ring at position 3 or the 4'-hydroxy or -methoxy substituent blocks increased SIRT1 expression.
Collapse
Affiliation(s)
- Kyle A Rasbach
- Medical University of South Carolina, Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, 280 Calhoun St., P.O. Box 250140, Charleston, SC 29425, USA
| | | |
Collapse
|
42
|
d-Serine exposure resulted in gene expression changes indicative of activation of fibrogenic pathways and down-regulation of energy metabolism and oxidative stress response. Toxicology 2008; 243:177-92. [DOI: 10.1016/j.tox.2007.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Revised: 09/18/2007] [Accepted: 10/08/2007] [Indexed: 11/21/2022]
|
43
|
Tsukamoto Y, Kotani T, Shiraishi Y, Kawamura H, Sakuma S. Epithelial cell proliferation of collecting ducts and ureters in the regenerating process of interstitial nephritis caused by infectious bronchitis virus. Avian Pathol 2007; 25:95-102. [DOI: 10.1080/03079459608419123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
44
|
Druml W. [Prevention and therapy of acute renal failure: the importance of metabolic intervention]. Wien Klin Wochenschr 2007; 119:201-4. [PMID: 17492345 DOI: 10.1007/s00508-007-0793-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
45
|
|
46
|
van Timmeren MM, Bakker SJL, Vaidya VS, Bailly V, Schuurs TA, Damman J, Stegeman CA, Bonventre JV, van Goor H. Tubular kidney injury molecule-1 in protein-overload nephropathy. Am J Physiol Renal Physiol 2006; 291:F456-64. [PMID: 16467126 DOI: 10.1152/ajprenal.00403.2005] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Kim-1, a recently discovered membrane protein, is undetectable in normal kidneys but markedly induced in proximal tubules after ischemic and toxic injury. The function of Kim-1 is unclear, but it is implicated in damage/repair processes. The Kim-1 ectodomain is cleaved by metalloproteinases and detectable in urine. We studied Kim-1 in a nontoxic, nonischemic, model of tubulointerstitial damage caused by acute proteinuria. Uninephrectomized (NX) rats received daily (ip) injections of 2 g BSA (NX+BSA, n = 12) or saline (NX, n = 6) for 3 wk. Kidneys were stained for various damage markers by immunohistochemistry (IHC). Kim-1 mRNA (RT-PCR, in situ hybridization), protein (IHC, Western blotting), and urinary Kim-1 (Luminex) were determined. Spatial relations between Kim-1 and other damage markers were studied by double labeling IHC. NX+BSA rats developed massive proteinuria (1,217 ± 313 vs. 18 ± 2 mg/day in NX, P < 0.001) and significant renal damage. Kim-1 mRNA was upregulated eightfold in NX+BSA (ratio Kim-1/β-actin, 4.08 ± 2.56 vs. 0.52 ± 0.64 in NX, P < 0.001) and localized to damaged tubules. Kim-1 protein expression was markedly induced in NX+BSA (2.46 ± 1.19 vs. 0.39 ± 0.10% staining/field in NX, P < 0.001). Urinary Kim-1 was significantly elevated in NX+BSA (921 ± 592 vs. 87 ± 164 pg/ml in NX, P < 0.001) and correlated with tissue Kim-1 expression ( r = 0.66, P =0.02). Kim-1 protein was found at the apical membrane of dilated nephrons. Kim-1 expression was limited to areas with inflammation (MØ), fibrosis (α-smooth muscle actin), and tubular damage (osteopontin), and only occasionally with tubular dedifferentiation (vimentin). These results implicate involvement of Kim-1 in the pathogenesis of proteinuria-induced renal damage/repair. Urinary Kim-1 levels may serve as a marker of proteinuria-induced renal damage.
Collapse
Affiliation(s)
- Mirjan M van Timmeren
- Department of Pathology and Laboratory Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kwon DS, Kwon CH, Kim JH, Woo JS, Jung JS, Kim YK. Signal transduction of MEK/ERK and PI3K/Akt activation by hypoxia/reoxygenation in renal epithelial cells. Eur J Cell Biol 2006; 85:1189-99. [PMID: 16860436 DOI: 10.1016/j.ejcb.2006.06.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Revised: 06/09/2006] [Accepted: 06/12/2006] [Indexed: 10/24/2022] Open
Abstract
The extracellular signal-regulated kinase (ERK) and Akt have been reported to be activated by ischemia/reperfusion in vivo. However, the signaling pathways involved in activation of these kinases and their potential roles were not fully understood in the postischemic kidney. In the present study, we observed that these kinases are activated by hypoxia/reoxygenation (H/R), an in vitro model of ischemia/reperfusion, in opossum kidney (OK) cells and elucidated the signaling pathways of these kinases. ERK and Akt were transiently activated during the early phase of reoxygenation following 4-12h of hypoxia. The ERK activation was inhibited by U0126, a specific inhibitor of ERK upstream MAPK/ERK kinase (MEK), but not by LY294002, a specific inhibitor of phosphoinositide 3-kinase (PI3K), whereas Akt activation was blocked by LY294002, but not by U0126. Inhibitors of epidermal growth factor receptor (EGFR) (AG 1478), Ras and Raf, as well as antioxidants inhibited activation of ERK and Akt, while the Src inhibitor PP2 had no effect. PI3K/Akt activation was shown to be associated with up-regulation of X chromosome-linked inhibitor of apoptosis (XIAP), but not survivin. Reoxygenation following 4-h hypoxia-stimulated cell proliferation, which was dependent on ERK and Akt activation and was also inhibited by antioxidants and AG 1478. Taken together, these results suggest that H/R induces activation of MEK/ERK and PI3K/Akt/XIAP survival signaling pathways through the reactive oxygen species-dependent EGFR/Ras/Raf cascade. Activation of these kinases may be involved in the repair process during ischemia/reperfusion.
Collapse
Affiliation(s)
- Dae Sik Kwon
- Department of Physiology, Medical Research Institute, MRC for Ischemic Tissue Regeneration, College of Medicine, Pusan National University, Pusan 602-739, Republic of Korea
| | | | | | | | | | | |
Collapse
|
48
|
Reuters I, Weber M, Schulze-Lohoff E. Rho/Rho kinase pathway regulates maintenance of the differentiated tubular epithelial cell phenotype on laminin-1. Nephron Clin Pract 2006; 104:p95-p106. [PMID: 16847378 DOI: 10.1159/000094573] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2005] [Accepted: 04/02/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Maintenance of a polarized tubular epithelium by appropriate intracellular signaling and extracellular matrix is critical both in normal renal function as well as in acute and chronic tubular injury. We examined the hypothesis that maintenance of a differentiated epithelial phenotype on the basement membrane glycoprotein laminin-1 is controlled by the Rho/Rho kinase pathway. METHODS Using the tubular epithelial cell lines LLC-PK1 and MDCK which were cultured on laminin-1 vs. collagen IV, we analyzed cell morphology and motility (cohort migration assay) as well as expression of differentiation and dedifferentiation markers (immunofluorescence microscopy). RESULTS Cohort migration of LLC-PK1 cells was significantly slowed down on laminin-1 (10.7 +/- 2.2 m.u. (migratory units)) compared with collagen IV (16.6 +/- 2.3 m.u.; BSA control: 2.8 +/- 2.5 m.u.). Inhibition of the Rho/Rho kinase pathway by C3 exotoxin (1 mug/ml) or the Rho kinase inhibitor Y27632 (10 microM) significantly augmented cohort migration on laminin-1 (14.5 +/- 1.4 and 16.0 +/- 1.8 m.u. vs. 10.7 +/- 2.2 m.u.). In parallel to the increased migratory activity, inhibition of the Rho/Rho kinase pathway resulted in a more mesenchymal phenotype of LLC-PK1 cells on laminin-1 with increased formation of lamellopodia and filopodia, distinct loss of focal contacts and stress fibers, upregulation of the dedifferentiation marker vimentin, and loss of cell-cell contacts with translocation of beta-catenin from the adherens junctions to the cytosol and nucleus. Similarly, cohort migration of MDCK cells was retarded on laminin-1 when compared with collagen IV, and addition of the Rho kinase inhibitor Y27632 resulted in enhanced motility and a change in cell morphology. CONCLUSION The study demonstrates that the Rho/Rho kinase pathway is required to maintain a non-migratory epithelial phenotype of cultured renal tubular LLC-PK1 and MDCK cells on the basement membrane glycoprotein laminin-1.
Collapse
Affiliation(s)
- Irith Reuters
- Department of Medicine I, Cologne General Hospital, Merheim Medical Center, Cologne, Germany
| | | | | |
Collapse
|
49
|
Cheng CW, Rifai A, Ka SM, Shui HA, Lin YF, Lee WH, Chen A. Calcium-binding proteins annexin A2 and S100A6 are sensors of tubular injury and recovery in acute renal failure. Kidney Int 2006; 68:2694-703. [PMID: 16316344 DOI: 10.1111/j.1523-1755.2005.00740.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Rise in cellular calcium is associated with acute tubular necrosis, the most common cause of acute renal failure (ARF). The mechanisms that calcium signaling induce in the quiescent tubular cells to proliferate and differentiate during acute tubular necrosis have not been elucidated. METHODS Acute tubular necrosis induced in mice by single intravenous injection of uranyl nitrate and examined after 1, 3, 7, and 14 days. Renal function was monitored and kidneys were evaluated by histology, immunohistochemistry, Western blotting, in situ hybridization, and real-time reverse transcription-polymerase chain reaction (RT-PCR). Models of folic acid induced-ARF and ischemic/reperfusion (I/R) injury were similarly investigated. RESULTS Analysis of mRNA expression of intracellular calcium and phospholipid-binding proteins demonstrated selective expression of S100A6 and Annexin A2 (Anxa2) in the renal cortex with marked elevation on day 3, and gradually decline on day 7 and further attenuation on day 14. Similarly, the expression of both proteins, as demonstrated by immunohistochemistry and Western blot analysis, was increased and reached the peak level on day 7 and then gradually declined by day 14. Vimentin, a marker of dedifferentiated cells, was highly expressed during the recovery phase. Combined in situ hybridization immunohistochemistry revealed colocalization of both S100A6 and Anxa2 with proliferating cell nuclear antigen (PCNA). The universality of this phenomenon was confirmed in two other mouse acute tubular necrosis models, the ischemic-reperfusion injury and folic acid-induced ARF. CONCLUSION Collectively, these findings demonstrate that S100A6 and Anxa2 expression, initiated in response to tubular injury, persist in parallel throughout the recovery process of tubular cells in acute renal failure.
Collapse
Affiliation(s)
- Chao-Wen Cheng
- Graduate Institute of Life Sciences, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
50
|
Gill N, Nally JV, Fatica RA. Renal failure secondary to acute tubular necrosis: epidemiology, diagnosis, and management. Chest 2005; 128:2847-63. [PMID: 16236963 DOI: 10.1378/chest.128.4.2847] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Acute tubular necrosis (ATN) is a form of acute renal failure (ARF) that is common in hospitalized patients. In critical care units, it accounts for about 76% of cases of ARF. Despite the introduction of hemodialysis > 30 years ago, the mortality rates from ATN in hospitalized and ICU patients are about 37.1% and 78.6%, respectively. The purpose of this review is to discuss briefly the cause, diagnosis, and epidemiology of ARF, and to review in depth the clinical trials performed to date that have examined the influence of growth factors, hormones, antioxidants, diuretics, and dialysis. In particular, the role of the dialysis modality, dialyzer characteristics, and dosing strategies are discussed.
Collapse
Affiliation(s)
- Namita Gill
- Department of General Internal Medicine, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA.
| | | | | |
Collapse
|