1
|
Jochmans I, Lerut E, Monbaliu D, Pirenne J. Impact of a Single Dose of Alpha-1-Antitrypsin in a Rat Model of Bilateral Kidney Ischemia Reperfusion Injury. J Surg Res 2024; 299:179-187. [PMID: 38759334 DOI: 10.1016/j.jss.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/19/2024]
Abstract
INTRODUCTION Renal ischemia reperfusion injury is a major cause of perioperative acute kidney injury. Alpha-1-antitrypsin (AAT), a protease inhibitor, might improve outcomes by reducing inflammation and apoptosis. We investigated the effects of a single intravenous dose of AAT immediately before ischemia in a rat bilateral renal clamping model. METHODS Both renal pedicles of male Sprague-Dawley rats were clamped (45 min). Plasma and renal tissue were collected at 3 h, 24 h, and 7 d. Intravenous AAT (60 mg/kg) was administered 5 min before clamping. Controls received saline. Shams underwent surgery without clamping or injection. Kidney function was assessed by plasma creatinine; injury by aspartate aminotransferase, heart-type-fatty-acid-binding-protein, and histopathology. Renal gene expression of tumor necrosis factor α, interleukin (IL)-6, heat shock protein 70, Chemokine (C-X-C motif) ligand 2, cyclo-oxygenase 2, endothelin-1, IL-10, heme oxygenase 1, B-cell lymphoma 2, and bcl-2-like protein 4 were determined by quantitative reverse transcriptase polymerase chain reaction. RESULTS None of the 3 h and 24 h end points were different between Control and AAT. In Sham, survival was 100% (6/6), 33% in Control (2/6), and 83% (5/6) in AAT (overall log-rank 0.03). At 7 d, plasma creatinine was lower with higher glomerular filtration rate in surviving AAT treated animals compared to Control (P < 0.001, P 0.03, respectively). These also had lower tumor necrosis factor α and IL-6 gene expression (P 0.001, P < 0.001, respectively). CONCLUSIONS These data suggest that a single intravenous dose of AAT immediately before ischemia might affect proinflammatory gene expression, glomerular filtration rate and animal survival at 1 wk after reperfusion despite an absence of improvement in early renal function and injury. These findings deserve further investigating in sufficiently powered studies including both sexes.
Collapse
Affiliation(s)
- Ina Jochmans
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| | - Evelyne Lerut
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Diethard Monbaliu
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Zhu W, Ou Y, Wang C, An R, Lai J, Shen Y, Ye X, Wang H. A neutrophil elastase inhibitor, sivelestat, attenuates sepsis-induced acute kidney injury by inhibiting oxidative stress. Heliyon 2024; 10:e29366. [PMID: 38638960 PMCID: PMC11024609 DOI: 10.1016/j.heliyon.2024.e29366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/20/2024] Open
Abstract
Background Sivelestat, a selective inhibitor of neutrophil elastase (NE), can mitigate sepsis-related acute lung injury. However, the role of sivelestat in inhibiting oxidative stress and attenuating sepsis-related acute kidney injury (AKI) remains unclear. Here, we reported the effects of sivelestat against oxidative stress-induced AKI by suppressing the production of oxidative stress indicators. Materials and methods A male Sprague-Dawley rat model of sepsis was established by cecal ligation and puncture (CLP). Sivelestat or normal saline was administered into jugular vein with a sustained-release drug delivery system. Indicators of inflammation and AKI, including white blood cells (WBC), neutrophils, lymphocytes, C-reactive proteins (CRP), procalcitonin (PCT), blood urea nitrogen (BUN), creatinine (Cr) and uric acid (UA), were assessed at 24 h post-sivelestat treatment. Indicators of liver injury, including direct bilirubin (DBIL), indirect bilirubin (IBIL), aspartate aminotransferase (AST) and alanine aminotransferase (ALT), were also assessed at 24 h post-sivelestat treatment. Indicators of oxidative stress, including superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px), were assessed at 12 h and 24 h post-sivelestat treatment. At 24 h post-sivelestat treatment, H&E staining of kidney and liver tissue was performed to observe pathological alterations. Results At 24 h post normal saline or sivelestat (0.2 g/kg body weight) treatment, WBC, neutrophil, CRP, PCT, MDA, BUN, Cr, UA, AST, ALT, DBIL and IBIL were increased, while SOD and GSH-Px were decreased, in septic rats treated with normal saline compared with that in non-septic rats treated with normal saline (all p < 0.05). The changes of these indicators were reversed in septic rats treated with sivelestat compared with that in septic rats treated with normal saline (all p < 0.05). Similar results were found regarding the levels of oxidative stress indicators at 12 h post-sivelestat treatment. The degenerative histopathological changes in both kidney and liver tissues were ameliorated upon sivelestat treatment. Conclusions Sivelestat plays a protective role in sepsis-related AKI by inhibiting oxidative stress. Our study reveals a possible therapeutic potential of sivelestat for oxidative stress-induced AKI.
Collapse
Affiliation(s)
- Wei Zhu
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Yingwei Ou
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Chunnian Wang
- Ningbo Clinical Pathology Diagnosis Center, Ningbo 315000, Zhejiang, China
| | - Rongcheng An
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Junmei Lai
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Ye Shen
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Xiangming Ye
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Haochu Wang
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
3
|
Jaimes Campos MA, Mavrogeorgis E, Latosinska A, Eder S, Buchwinkler L, Mischak H, Siwy J, Rossing P, Mayer G, Jankowski J. Urinary peptide analysis to predict the response to blood pressure medication. Nephrol Dial Transplant 2024; 39:873-883. [PMID: 37930730 PMCID: PMC11181870 DOI: 10.1093/ndt/gfad223] [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: 06/17/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The risk of diabetic kidney disease (DKD) progression is significant despite treatment with renin-angiotensin system (RAS) blocking agents. Current clinical tools cannot predict whether or not patients will respond to treatment with RAS inhibitors (RASi). We aimed to investigate whether proteome analysis could identify urinary peptides as biomarkers that could predict the response to angiotensin-converting enzyme inhibitor and angiotensin-receptor blockers treatment to avoid DKD progression. Furthermore, we investigated the comparability of the estimated glomerular filtration rate (eGFR), calculated using four different GFR equations, for DKD progression. METHODS We evaluated urine samples from a discovery cohort of 199 diabetic patients treated with RASi. DKD progression was defined based on eGFR percentage slope results between visits (∼1 year) and for the entire period (∼3 years) based on the eGFR values of each GFR equation. Urine samples were analysed using capillary electrophoresis-coupled mass spectrometry. Statistical analysis was performed between the uncontrolled (patients who did not respond to RASi treatment) and controlled kidney function groups (patients who responded to the RASi treatment). Peptides were combined in a support vector machine-based model. The area under the receiver operating characteristic curve was used to evaluate the risk prediction models in two independent validation cohorts treated with RASi. RESULTS The classification of patients into uncontrolled and controlled kidney function varies depending on the GFR equation used, despite the same sample set. We identified 227 peptides showing nominal significant difference and consistent fold changes between uncontrolled and controlled patients in at least three methods of eGFR calculation. These included fragments of collagens, alpha-1-antitrypsin, antithrombin-III, CD99 antigen and uromodulin. A model based on 189 of 227 peptides (DKDp189) showed a significant prediction of non-response to the treatment/DKD progression in two independent cohorts. CONCLUSIONS The DKDp189 model demonstrates potential as a predictive tool for guiding treatment with RASi in diabetic patients.
Collapse
Affiliation(s)
- Mayra Alejandra Jaimes Campos
- Mosaiques Diagnostics GmbH, Hannover, Germany
- University Hospital RWTH Aachen, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | - Emmanouil Mavrogeorgis
- Mosaiques Diagnostics GmbH, Hannover, Germany
- University Hospital RWTH Aachen, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | | | - Susanne Eder
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Lukas Buchwinkler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Complications Research, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Joachim Jankowski
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
4
|
Pejchinovski I, Turkkan S, Pejchinovski M. Recent Advances of Proteomics in Management of Acute Kidney Injury. Diagnostics (Basel) 2023; 13:2648. [PMID: 37627907 PMCID: PMC10453063 DOI: 10.3390/diagnostics13162648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Acute Kidney Injury (AKI) is currently recognized as a life-threatening disease, leading to an exponential increase in morbidity and mortality worldwide. At present, AKI is characterized by a significant increase in serum creatinine (SCr) levels, typically followed by a sudden drop in glomerulus filtration rate (GFR). Changes in urine output are usually associated with the renal inability to excrete urea and other nitrogenous waste products, causing extracellular volume and electrolyte imbalances. Several molecular mechanisms were proposed to be affiliated with AKI development and progression, ultimately involving renal epithelium tubular cell-cycle arrest, inflammation, mitochondrial dysfunction, the inability to recover and regenerate proximal tubules, and impaired endothelial function. Diagnosis and prognosis using state-of-the-art clinical markers are often late and provide poor outcomes at disease onset. Inappropriate clinical assessment is a strong disease contributor, actively driving progression towards end stage renal disease (ESRD). Proteins, as the main functional and structural unit of the cell, provide the opportunity to monitor the disease on a molecular level. Changes in the proteomic profiles are pivotal for the expression of molecular pathways and disease pathogenesis. Introduction of highly-sensitive and innovative technology enabled the discovery of novel biomarkers for improved risk stratification, better and more cost-effective medical care for the ill patients and advanced personalized medicine. In line with those strategies, this review provides and discusses the latest findings of proteomic-based biomarkers and their prospective clinical application for AKI management.
Collapse
Affiliation(s)
- Ilinka Pejchinovski
- Department of Quality Assurance, Nikkiso Europe GmbH, 30885 Langenhagen, Germany; (I.P.); (S.T.)
| | - Sibel Turkkan
- Department of Quality Assurance, Nikkiso Europe GmbH, 30885 Langenhagen, Germany; (I.P.); (S.T.)
| | - Martin Pejchinovski
- Department of Analytical Instruments Group, Thermo Fisher Scientific, 82110 Germering, Germany
| |
Collapse
|
5
|
Bai Y, Li Y, Tang Z, Hu L, Jiang X, Chen J, Huang S, Wu K, Xu W, Chen C. Urinary proteome analysis of acute kidney injury in post-cardiac surgery patients using enrichment materials with high-resolution mass spectrometry. Front Bioeng Biotechnol 2022; 10:1002853. [PMID: 36177176 PMCID: PMC9513377 DOI: 10.3389/fbioe.2022.1002853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Cardiac surgery-associated acute kidney injury (CSA-AKI) may increase the mortality and incidence rates of chronic kidney disease in critically ill patients. This study aimed to investigate the underlying correlations between urinary proteomic changes and CSA-AKI. Methods: Nontargeted proteomics was performed using nano liquid chromatography coupled with Orbitrap Exploris mass spectrometry (MS) on urinary samples preoperatively and postoperatively collected from patients with CSA-AKI. Gemini C18 silica microspheres were used to separate and enrich trypsin-hydrolysed peptides under basic mobile phase conditions. Differential analysis was conducted to screen out urinary differential expressed proteins (DEPs) among patients with CSA-AKI for bioinformatics. Kyoto Encyclopedia of Genes and Genomes (KEGG) database analysis was adopted to identify the altered signal pathways associated with CSA-AKI. Results: Approximately 2000 urinary proteins were identified and quantified through data-independent acquisition MS, and 324 DEPs associated with AKI were screened by univariate statistics. According to KEGG enrichment analysis, the signal pathway of protein processing in the endoplasmic reticulum was enriched as the most up-regulated DEPs, and cell adhesion molecules were enriched as the most down-regulated DEPs. In protein–protein interaction analysis, the three hub targets in the up-regulated DEPs were α-1-antitrypsin, β-2-microglobulin and angiotensinogen, and the three key down-regulated DEPs were growth arrest-specific protein 6, matrix metalloproteinase-9 and urokinase-type plasminogen activator. Conclusion: Urinary protein disorder was observed in CSA-AKI due to ischaemia and reperfusion. The application of Gemini C18 silica microspheres can improve the protein identification rate to obtain highly valuable resources for the urinary DEPs of AKI. This work provides valuable knowledge about urinary proteome biomarkers and essential resources for further research on AKI.
Collapse
Affiliation(s)
- Yunpeng Bai
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Department of Critical Care Medicine, Maoming People’s Hospital, Maoming, China
| | - Ying Li
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhizhong Tang
- Department of Urology, Maoming People’s Hospital, Maoming, China
| | - Linhui Hu
- Department of Critical Care Medicine, Maoming People’s Hospital, Maoming, China
| | - Xinyi Jiang
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jingchun Chen
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Sumei Huang
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Department of Emergency, Maoming People’s Hospital, Maoming, China
- Biological Resource Center of Maoming People’s Hospital, Maoming, China
| | - Kunyong Wu
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Biological Resource Center of Maoming People’s Hospital, Maoming, China
| | - Wang Xu
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chunbo Chen
- Department of Emergency, Maoming People’s Hospital, Maoming, China
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
- *Correspondence: Chunbo Chen, , orcid.org/0000-0001-5662-497X
| |
Collapse
|
6
|
Ong KL, Wu L, Januszewski AS, O’Connell R, Xu A, Scott RS, Sullivan DR, Rye KA, Li H, Ma RCW, Li L, Gebski V, Jenkins AJ, Jia W, Keech AC. The relationship of neutrophil elastase and proteinase 3 with risk factors, and chronic complications in type 2 diabetes: A Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) sub-study. Diab Vasc Dis Res 2021; 18:14791641211032547. [PMID: 34275349 PMCID: PMC8481737 DOI: 10.1177/14791641211032547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Neutrophil elastase (NE) and proteinase 3 (PR3) are novel inflammation biomarkers. We investigated their associations with chronic complications, determinants of biomarker levels and effects of fenofibrate in patients with type 2 diabetes mellitus (T2DM) from Fenofibrate Intervention and Event Lowering in Diabetes study. METHODS Plasma NE and PR3 levels were quantified at baseline (n = 2000), and relationships with complications over 5-years assessed. Effects of fenofibrate on biomarker levels (n = 200) were determined at four follow-up visits. RESULTS Higher waist-to-hip ratio, homocysteine and C-reactive protein and lower apoA-II were determinants of higher NE and PR3 levels. Higher NE levels were associated with on-trial stroke and cardiovascular mortality, and higher PR3 levels with on-trial stroke, but associations were not significant after adjustment for confounding factors. Although higher NE and PR3 levels were associated with baseline total microvascular disease, only NE levels were associated with on-trial neuropathy or amputation. These associations were not significant after adjusting for multiple comparisons. NE and PR3 levels did not change with fenofibrate. CONCLUSIONS In T2DM plasma NE and PR3 levels are associated with vascular risk factors, and total microvascular disease at baseline, but on rigorous analyses were not associated with on-trial complications. Levels were not changed by fenofibrate.
Collapse
Affiliation(s)
- Kwok-Leung Ong
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Liang Wu
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | | | - Rachel O’Connell
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
| | - Russell S Scott
- Lipid and Diabetes Research Group, Christchurch Hospital, Christchurch, New Zealand
| | - David R Sullivan
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Chemical Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ronald CW Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Liping Li
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Val Gebski
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Alicia J Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Anthony C Keech
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Prof Anthony Keech, NHMRC Clinical Trials Centre, Level 6, Medical Foundation Building, 92–94 Parramatta Road, Camperdown, NSW 2050, Australia.
| |
Collapse
|
7
|
Ostermann L, Maus R, Stolper J, Schütte L, Katsarou K, Tumpara S, Pich A, Mueller C, Janciauskiene S, Welte T, Maus UA. Alpha-1 antitrypsin deficiency impairs lung antibacterial immunity in mice. JCI Insight 2021; 6:140816. [PMID: 33554955 PMCID: PMC7934856 DOI: 10.1172/jci.insight.140816] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) is a major inhibitor of serine proteases in mammals. Therefore, its deficiency leads to protease–antiprotease imbalance and a risk for developing lung emphysema. Although therapy with human plasma-purified AAT attenuates AAT deficiency–related emphysema, its impact on lung antibacterial immunity is poorly defined. Here, we examined the effect of AAT therapy on lung protective immunity in AAT-deficient (KO) mice challenged with Streptococcus pneumoniae. AAT-KO mice were highly susceptible to S. pneumoniae, as determined by severe lobar pneumonia and early mortality. Mechanistically, we found that neutrophil-derived elastase (NE) degraded the opsonophagocytically important collectins, surfactant protein A (SP-A) and D (SP-D), which was accompanied by significantly impaired lung bacterial clearance in S. pneumoniae–infected AAT-KO mice. Treatment of S. pneumoniae–infected AAT-KO mice with human AAT protected SP-A and SP-D from NE-mediated degradation and corrected the pulmonary pathology observed in these mice. Likewise, treatment with Sivelestat, a specific inhibitor of NE, also protected collectins from degradation and significantly decreased bacterial loads in S. pneumoniae–infected AAT-KO mice. Our findings show that NE is responsible for the degradation of lung SP-A and SP-D in AAT-KO mice affecting lung protective immunity in AAT deficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andreas Pich
- Institute of Toxicology and Core Facility Proteomics, Hannover Medical School, Hannover, Germany
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sabina Janciauskiene
- Clinic for Pneumology, and.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Tobias Welte
- Clinic for Pneumology, and.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Ulrich A Maus
- Division of Experimental Pneumology.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| |
Collapse
|
8
|
Róka B, Tod P, Kaucsár T, Vizovišek M, Vidmar R, Turk B, Fonović M, Szénási G, Hamar P. The Acute Phase Response Is a Prominent Renal Proteome Change in Sepsis in Mice. Int J Mol Sci 2019; 21:E200. [PMID: 31892161 PMCID: PMC6982205 DOI: 10.3390/ijms21010200] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
(1) Background: Sepsis-induced acute kidney injury (AKI) is the most common form of acute kidney injury (AKI). We studied the temporal profile of the sepsis-induced renal proteome changes. (2) Methods: Male mice were injected intraperitoneally with bacterial lipopolysaccharide (LPS) or saline (control). Renal proteome was studied by LC-MS/MS (ProteomeXchange: PXD014664) at the early phase (EP, 1.5 and 6 h after 40 mg/kg LPS) and the late phase (LP, 24 and 48 h after 10 mg/kg LPS) of LPS-induced AKI. Renal mRNA expression of acute phase proteins (APP) was assessed by qPCR. (3) Results: Renal proteome change was milder in EP vs. LP. APPs dominated the proteome in LP (proteins upregulated at least 4-fold (APPs/all): EP, 1.5 h: 0/10, 6 h: 1/10; LP, 24 h: 22/47, 48 h: 17/44). Lipocalin-2, complement C3, fibrinogen, haptoglobin and hemopexin were the most upregulated APPs. Renal mRNA expression preceded the APP changes with peak effects at 24 h, and indicated renal production of the majority of APPs. (4) Conclusions: Gene expression analysis revealed local production of APPs that commenced a few hours post injection and peaked at 24 h. This is the first demonstration of a massive, complex and coordinated acute phase response of the kidney involving several proteins not identified previously.
Collapse
Affiliation(s)
- Beáta Róka
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
| | - Pál Tod
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kaucsár
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
| | - Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
| | - Robert Vidmar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
- Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, 1000 Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
- Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, 1000 Ljubljana, Slovenia
| | - Gábor Szénási
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
9
|
Taherkhani A, Farrokhi Yekta R, Mohseni M, Saidijam M, Arefi Oskouie A. Chronic kidney disease: a review of proteomic and metabolomic approaches to membranous glomerulonephritis, focal segmental glomerulosclerosis, and IgA nephropathy biomarkers. Proteome Sci 2019; 17:7. [PMID: 31889913 PMCID: PMC6925425 DOI: 10.1186/s12953-019-0155-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic Kidney Disease (CKD) is a global health problem annually affecting millions of people around the world. It is a comprehensive syndrome, and various factors may contribute to its occurrence. In this study, it was attempted to provide an accurate definition of chronic kidney disease; followed by focusing and discussing on molecular pathogenesis, novel diagnosis approaches based on biomarkers, recent effective antigens and new therapeutic procedures related to high-risk chronic kidney disease such as membranous glomerulonephritis, focal segmental glomerulosclerosis, and IgA nephropathy, which may lead to end-stage renal diseases. Additionally, a considerable number of metabolites and proteins that have previously been discovered and recommended as potential biomarkers of various CKDs using ‘-omics-’ technologies, proteomics, and metabolomics were reviewed.
Collapse
Affiliation(s)
- Amir Taherkhani
- 1Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Maede Mohseni
- 3Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- 1Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Afsaneh Arefi Oskouie
- 4Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Turnier JL, Brunner HI, Bennett M, Aleed A, Gulati G, Haffey WD, Thornton S, Wagner M, Devarajan P, Witte D, Greis KD, Aronow B. Discovery of SERPINA3 as a candidate urinary biomarker of lupus nephritis activity. Rheumatology (Oxford) 2019; 58:321-330. [PMID: 30285245 DOI: 10.1093/rheumatology/key301] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Indexed: 12/11/2022] Open
Abstract
Objectives We used an unbiased proteomics approach to identify candidate urine biomarkers (CUBMs) predictive of LN chronicity and pursued their validation in a larger cohort. Methods In this cross-sectional pilot study, we selected urine collected at kidney biopsy from 20 children with varying levels of LN damage (discovery cohort) and performed proteomic analysis using isobaric tags for relative and absolute quantification (iTRAQ). We identified differentially excreted proteins based on degree of LN chronicity and sought to distinguish markers exhibiting different relative expression patterns using hierarchically clustered log10-normalized relative abundance data with linked and distinct functions by biological network analyses. For each CUBM, we performed specific ELISAs on urine from a validation cohort (n = 41) and analysis of variance to detect differences between LN chronicity, with LN activity adjustment. We evaluated for CUBM expression in LN biopsies with immunohistochemistry. Results iTRAQ detected 112 proteins in urine from the discovery cohort, 51 quantifiable in all replicates. Simple analysis of variance revealed four differentially expressed, chronicity-correlated proteins (P-values < 0.05). Further correlation and network analyses led to selection of seven CUBMs for LN chronicity. In the validation cohort, none of the CUBMs distinguished LN chronicity degree; however, urine SERPINA3 demonstrated a moderate positive correlation with LN histological activity. Immunohistochemistry further demonstrated SERPINA3 staining in proximal tubular epithelial and endothelial cells. Conclusion We identified SERPINA3, a known inhibitor of neutrophil cathepsin G and angiotensin II production, as a potential urine biomarker to help quantify LN activity.
Collapse
Affiliation(s)
- Jessica L Turnier
- Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hermine I Brunner
- Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael Bennett
- Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashwaq Aleed
- Department of Pediatrics, Qassim University College of Medicine, Qassim, Saudi Arabia
| | - Gaurav Gulati
- Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy D Haffey
- Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sherry Thornton
- Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael Wagner
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Prasad Devarajan
- Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David Witte
- Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D Greis
- Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bruce Aronow
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
11
|
Lee CW, Kou HW, Chou HS, Chou HH, Huang SF, Chang CH, Wu CH, Yu MC, Tsai HI. A combination of SOFA score and biomarkers gives a better prediction of septic AKI and in-hospital mortality in critically ill surgical patients: a pilot study. World J Emerg Surg 2018; 13:41. [PMID: 30214469 PMCID: PMC6131912 DOI: 10.1186/s13017-018-0202-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Background Sepsis is a syndrome characterized by a constellation of clinical manifestations and a significantly high mortality rate in the surgical intensive care unit (ICU). It is frequently complicated by acute kidney injury (AKI), which, in turn, increases the risk of mortality. Therefore, it is of paramount importance to identify those septic patients at risk for the development of AKI and mortality. The objective of this pilot study was to evaluate several different biomarkers, including NGAL, calprotectin, KIM-1, cystatin C, and GDF-15, along with SOFA scores, in predicting the development of septic AKI and associated in-hospital mortality in critically ill surgical patients. Methods Patients admitted to the surgical ICU were prospectively enrolled, having given signed informed consent. Their blood and urine samples were obtained and subjected to enzyme-linked immunosorbent assay (ELISA) to determine the levels of various novel biomarkers. The clinical data and survival outcome were recorded and analyzed. Results A total of 33 patients were enrolled in the study. Most patients received surgery prior to ICU admission, with abdominal surgery being the most common type of procedure (27 patients (81.8%)). In the study, 22 patients had a diagnosis of sepsis with varying degrees of AKI, while the remaining 11 were free of sepsis. Statistical analysis demonstrated that in patients with septic AKI versus those without, the following were significantly higher: serum NGAL (447.5 ± 35.7 ng/mL vs. 256.5 ± 31.8 ng/mL, P value 0.001), calprotectin (1030.3 ± 298.6 pg/mL vs. 248.1 ± 210.7 pg/mL, P value 0.049), urinary NGAL (434.2 ± 31.5 ng/mL vs. 208.3 ± 39.5 ng/mL, P value < 0.001), and SOFA score (11.5 ± 1.2 vs. 4.4 ± 0.5, P value < 0.001). On the other hand, serum NGAL (428.2 ± 32.3 ng/mL vs. 300.4 ± 44.3 ng/mL, P value 0.029) and urinary NGAL (422.3 ± 33.7 ng/mL vs. 230.8 ± 42.2 ng/mL, P value 0.001), together with SOFA scores (10.6 ± 1.4 vs. 5.6 ± 0.8, P value 0.003), were statistically higher in cases of in-hospital mortality. A combination of serum NGAL, urinary NGAL, and SOFA scores could predict in-hospital mortality with an AUROC of 0.911. Conclusions This pilot study demonstrated a promising panel that allows an early diagnosis, high sensitivity, and specificity and a prognostic value for septic AKI and in-hospital mortality in surgical ICU. Further study is warranted to validate our findings. Electronic supplementary material The online version of this article (10.1186/s13017-018-0202-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chao-Wei Lee
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China.,2College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan, Republic of China.,3Graduate Institute of Clinical Medical Sciences, Chang Gung University, Guishan, Taoyuan, Taiwan, Republic of China
| | - Hao-Wei Kou
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| | - Hong-Shiue Chou
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| | - Hsu-Huan Chou
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| | - Song-Fong Huang
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| | - Chih-Hsiang Chang
- Division of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| | - Chun-Hsing Wu
- 3Graduate Institute of Clinical Medical Sciences, Chang Gung University, Guishan, Taoyuan, Taiwan, Republic of China
| | - Ming-Chin Yu
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China.,2College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan, Republic of China
| | - Hsin-I Tsai
- 3Graduate Institute of Clinical Medical Sciences, Chang Gung University, Guishan, Taoyuan, Taiwan, Republic of China.,Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taiwan, Republic of China
| |
Collapse
|
12
|
Pontillo C, Jacobs L, Staessen JA, Schanstra JP, Rossing P, Heerspink HJL, Siwy J, Mullen W, Vlahou A, Mischak H, Vanholder R, Zürbig P, Jankowski J. A urinary proteome-based classifier for the early detection of decline in glomerular filtration. Nephrol Dial Transplant 2018; 32:1510-1516. [PMID: 27387473 DOI: 10.1093/ndt/gfw239] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/02/2016] [Indexed: 12/13/2022] Open
Abstract
Background Chronic kidney disease (CKD) progression is currently assessed by a decline in estimated glomerular filtration rate (eGFR) and/or an increase in urinary albumin excretion (UAE). However, these markers are considered either to be late-stage markers or to have low sensitivity or specificity. In this study, we investigated the performance of the urinary proteome-based classifier CKD273, compared with UAE, in a number of different narrow ranges of CKD severity, with each range separated by an eGFR of 10 mL/min/1.73 m 2 . Methods A total of 2672 patients with different CKD stages were included in the study. Of these, 394 individuals displayed a decline in eGFR of >5 mL/min/1.73 m 2 /year (progressors) and the remaining individuals were considered non-progressors. For all samples, UAE values and CKD273 classification scores were obtained. To assess UAE values and CKD273 scores at different disease stages, the cohort was divided according to baseline eGFRs of ≥80, 70-79, 60-69, 50-59, 40-49, 30-39 and <29 mL/min/1.73 m 2 . In addition, areas under the curve for CKD273 and UAE were calculated. Results In early stage CKD, the urinary proteome-based classifier performed significantly better than UAE in detecting progressors. In contrast, UAE performed better in patients with late-stage CKD. No significant difference in performance was found between CKD273 and UAE in patients with moderately reduced renal function. Conclusions These results suggest that urinary peptides, as combined in the CKD273 classifier, allow the detection of progressive CKD at early stages, a point where therapeutic intervention is more likely to be effective. However, late-stage disease, where irreversible damage of the kidney is already present, is better detected by UAE.
Collapse
Affiliation(s)
- Claudia Pontillo
- Mosaiques Diagnostics, Hanover, Germany.,Charité-Universitatsmedizin, Berlin, Germany
| | - Lotte Jacobs
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jan A Staessen
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.,R&D VitaK Group, Maastricht University, Maastricht, The Netherlands
| | - Joost P Schanstra
- Institute of Metabolic and Cardiovascular Diseases, Inserm U1048, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Peter Rossing
- Steno Diabetes Center, Gentofte, Denmark.,University of Aarhus, Aarhus, Denmark.,Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Harald Mischak
- Mosaiques Diagnostics, Hanover, Germany.,University of Glasgow, Glasgow, UK
| | - Ray Vanholder
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Joachim Jankowski
- Charité-Universitatsmedizin, Berlin, Germany.,Institute for Molecular Cardiovascular Research, University Hospital RWTH, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| |
Collapse
|
13
|
Lin H, Chen M, Tian F, Tikkanen J, Ding L, Andrew Cheung HY, Nakajima D, Wang Z, Mariscal A, Hwang D, Cypel M, Keshavjee S, Liu M. α 1 -Anti-trypsin improves function of porcine donor lungs during ex-vivo lung perfusion. J Heart Lung Transplant 2018; 37:656-666. [DOI: 10.1016/j.healun.2017.09.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 09/15/2017] [Accepted: 09/26/2017] [Indexed: 11/28/2022] Open
|
14
|
Carvalho MOS, Souza ALCS, Carvalho MB, Pacheco APAS, Rocha LC, do Nascimento VML, Figueiredo CVB, Guarda CC, Santiago RP, Adekile A, Goncalves MDS. Evaluation of Alpha-1 Antitrypsin Levels and SERPINA1 Gene Polymorphisms in Sickle Cell Disease. Front Immunol 2017; 8:1491. [PMID: 29163550 PMCID: PMC5681845 DOI: 10.3389/fimmu.2017.01491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022] Open
Abstract
Alpha-1 antitrypsin (AAT) is an inhibitor of neutrophil elastase and a member of the serine proteinase inhibitor (serpin) superfamily, and little is known about its activity in sickle cell disease (SCD). We hypothesize that AAT may undergo changes in SCD because of the high oxidative stress and inflammation associated with the disease. We have found high AAT levels in SCD patients compared to controls, while mutant genotypes of SERPINA1 gene had decreased AAT levels, in both groups. AAT showed negative correlation with red blood cells, hemoglobin (Hb), hematocrit, high-density lipoprotein cholesterol, urea, creatinine, and albumin and was positively correlated with mean corpuscular Hb concentration, white blood cells, neutrophils, Hb S, bilirubin, lactate dehydrogenase, ferritin, and C-reactive protein. Patients with higher levels of AAT had more infection episodes (OR = 1.71, CI: 1.05–2.65, p = 0.02), gallstones (OR = 1.75, CI: 1.03–2.97, p = 0.02), and had more blood transfusions (OR = 2.35, CI: 1.51–3.65, p = 0.0001). Our data on AAT association with laboratory indices of hemolysis and inflammation suggest that it may be positively associated with SCD severity; the negative correlations with renal parameters suggest a cytoprotective mechanism in SCD patients. In summary, AAT may need to be included in studies related to SCD and in the discussion of further therapeutic strategies.
Collapse
Affiliation(s)
- Magda Oliveira Seixas Carvalho
- Instituto Gonçalo Moniz-Fiocruz-Bahia (IGM-FIOCRUZ-Ba), Salvador, Brazil.,Complexo Hospitalar Universitário Professor Edgard Santos, Salvador, Brazil
| | | | | | | | | | | | - Camylla Vilas Boas Figueiredo
- Instituto Gonçalo Moniz-Fiocruz-Bahia (IGM-FIOCRUZ-Ba), Salvador, Brazil.,Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Caroline Conceição Guarda
- Instituto Gonçalo Moniz-Fiocruz-Bahia (IGM-FIOCRUZ-Ba), Salvador, Brazil.,Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Rayra Pereira Santiago
- Instituto Gonçalo Moniz-Fiocruz-Bahia (IGM-FIOCRUZ-Ba), Salvador, Brazil.,Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Adekunle Adekile
- Department of Pediatrics, Kuwait University, Kuwait City, Kuwait
| | - Marilda de Souza Goncalves
- Instituto Gonçalo Moniz-Fiocruz-Bahia (IGM-FIOCRUZ-Ba), Salvador, Brazil.,Universidade Federal da Bahia (UFBA), Salvador, Brazil
| |
Collapse
|
15
|
Trivedi P, Kumar RK, Iyer A, Boswell S, Gerarduzzi C, Dadhania VP, Herbert Z, Joshi N, Luyendyk JP, Humphreys BD, Vaidya VS. Targeting Phospholipase D4 Attenuates Kidney Fibrosis. J Am Soc Nephrol 2017; 28:3579-3589. [PMID: 28814511 DOI: 10.1681/asn.2016111222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 07/11/2017] [Indexed: 01/13/2023] Open
Abstract
Phospholipase D4 (PLD4), a single-pass transmembrane glycoprotein, is among the most highly upregulated genes in murine kidneys subjected to chronic progressive fibrosis, but the function of PLD4 in this process is unknown. Here, we found PLD4 to be overexpressed in the proximal and distal tubular epithelial cells of murine and human kidneys after fibrosis. Genetic silencing of PLD4, either globally or conditionally in proximal tubular epithelial cells, protected mice from the development of fibrosis. Mechanistically, global knockout of PLD4 modulated innate and adaptive immune responses and attenuated the upregulation of the TGF-β signaling pathway and α1-antitrypsin protein (a serine protease inhibitor) expression and downregulation of neutrophil elastase (NE) expression induced by obstructive injury. In vitro, treatment with NE attenuated TGF-β-induced accumulation of fibrotic markers. Furthermore, therapeutic targeting of PLD4 using specific siRNA protected mice from folic acid-induced kidney fibrosis and inhibited the increase in TGF-β signaling, decrease in NE expression, and upregulation of mitogen-activated protein kinase signaling. Immunoprecipitation/mass spectrometry and coimmunoprecipitation experiments confirmed that PLD4 binds three proteins that interact with neurotrophic receptor tyrosine kinase 1, a receptor also known as TrkA that upregulates mitogen-activated protein kinase. PLD4 inhibition also prevented the folic acid-induced upregulation of this receptor in mouse kidneys. These results suggest inhibition of PLD4 as a novel therapeutic strategy to activate protease-mediated degradation of extracellular matrix and reverse fibrosis.
Collapse
Affiliation(s)
- Priyanka Trivedi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ramya K Kumar
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ashwin Iyer
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sarah Boswell
- Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Casimiro Gerarduzzi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Vivekkumar P Dadhania
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Zach Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nikita Joshi
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Benjamin D Humphreys
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri; and
| | - Vishal S Vaidya
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; .,Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
16
|
Nicolaou O, Kousios A, Hadjisavvas A, Lauwerys B, Sokratous K, Kyriacou K. Biomarkers of systemic lupus erythematosus identified using mass spectrometry-based proteomics: a systematic review. J Cell Mol Med 2017; 21:993-1012. [PMID: 27878954 PMCID: PMC5387176 DOI: 10.1111/jcmm.13031] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022] Open
Abstract
Advances in mass spectrometry technologies have created new opportunities for discovering novel protein biomarkers in systemic lupus erythematosus (SLE). We performed a systematic review of published reports on proteomic biomarkers identified in SLE patients using mass spectrometry-based proteomics and highlight their potential disease association and clinical utility. Two electronic databases, MEDLINE and EMBASE, were systematically searched up to July 2015. The methodological quality of studies included in the review was performed according to Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Twenty-five studies were included in the review, identifying 241 SLE candidate proteomic biomarkers related to various aspects of the disease including disease diagnosis and activity or pinpointing specific organ involvement. Furthermore, 13 of the 25 studies validated their results for a selected number of biomarkers in an independent cohort, resulting in the validation of 28 candidate biomarkers. It is noteworthy that 11 candidate biomarkers were identified in more than one study. A significant number of potential proteomic biomarkers that are related to a number of aspects of SLE have been identified using mass spectrometry proteomic approaches. However, further studies are required to assess the utility of these biomarkers in routine clinical practice.
Collapse
Affiliation(s)
- Orthodoxia Nicolaou
- Department of Electron Microscopy/Molecular PathologyThe Cyprus Institute of Neurology and GeneticsNicosiaCyprus
- Department of Electron Microscopy/Molecular PathologyCyprus School of Molecular MedicineNicosiaCyprus
| | - Andreas Kousios
- Department of Electron Microscopy/Molecular PathologyCyprus School of Molecular MedicineNicosiaCyprus
| | - Andreas Hadjisavvas
- Department of Electron Microscopy/Molecular PathologyThe Cyprus Institute of Neurology and GeneticsNicosiaCyprus
- Department of Electron Microscopy/Molecular PathologyCyprus School of Molecular MedicineNicosiaCyprus
| | - Bernard Lauwerys
- Department of RheumatologyUniversité catholique de LouvainBruxellesBelgium
| | - Kleitos Sokratous
- Department of Electron Microscopy/Molecular PathologyThe Cyprus Institute of Neurology and GeneticsNicosiaCyprus
| | - Kyriacos Kyriacou
- Department of Electron Microscopy/Molecular PathologyThe Cyprus Institute of Neurology and GeneticsNicosiaCyprus
- Department of Electron Microscopy/Molecular PathologyCyprus School of Molecular MedicineNicosiaCyprus
| |
Collapse
|
17
|
Maicas N, van der Vlag J, Bublitz J, Florquin S, Bakker-van Bebber M, Dinarello CA, Verweij V, Masereeuw R, Joosten LA, Hilbrands LB. Human Alpha-1-Antitrypsin (hAAT) therapy reduces renal dysfunction and acute tubular necrosis in a murine model of bilateral kidney ischemia-reperfusion injury. PLoS One 2017; 12:e0168981. [PMID: 28235038 PMCID: PMC5325207 DOI: 10.1371/journal.pone.0168981] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/11/2016] [Indexed: 11/18/2022] Open
Abstract
Several lines of evidence have demonstrated the anti-inflammatory and cytoprotective effects of alpha-1-antitrypsin (AAT), the major serum serine protease inhibitor. The aim of the present study was to investigate the effects of human AAT (hAAT) monotherapy during the early and recovery phase of ischemia-induced acute kidney injury. Mild renal ischemia-reperfusion (I/R) injury was induced in male C57Bl/6 mice by bilateral clamping of the renal artery and vein for 20 min. hAAT (80 mg/kg, Prolastin®) was administered daily intraperitoneally (i.p.) from day -1 until day 7 after surgery. Control animals received the same amount of human serum albumin (hAlb). Plasma, urine and kidneys were collected at 2h, 1, 2, 3, 8 and 15 days after reperfusion for histological and biochemical analysis. hAAT partially preserved renal function and tubular integrity after induction of bilateral kidney I/R injury, which was accompanied with reduced renal influx of macrophages and a significant decrease of neutrophil gelatinase-associated lipocalin (NGAL) protein levels in urine and plasma. During the recovery phase, hAAT significantly decreased kidney injury molecule-1 (KIM-1) protein levels in urine but showed no significant effect on renal fibrosis. Although the observed effect size of hAAT administration was limited and therefore the clinical relevance of our findings should be evaluated carefully, these data support the potential of this natural protein to ameliorate ischemic and inflammatory conditions.
Collapse
Affiliation(s)
- Nuria Maicas
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Janin Bublitz
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sandrine Florquin
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medicine, University of Colorado Health Sciences Center Denver, Colorado, United States of America
| | - Vivienne Verweij
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Roos Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - Leo A Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Pérez V, López D, Boixadera E, Ibernón M, Espinal A, Bonet J, Romero R. Comparative differential proteomic analysis of minimal change disease and focal segmental glomerulosclerosis. BMC Nephrol 2017; 18:49. [PMID: 28158993 PMCID: PMC5291957 DOI: 10.1186/s12882-017-0452-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/16/2017] [Indexed: 01/18/2023] Open
Abstract
Background Minimal change disease (MCD) and primary focal segmental glomerulosclerosis (FSGS) are glomerular diseases characterized by nephrotic syndrome. Their diagnosis requires a renal biopsy, but it is an invasive procedure with potential complications. In a small biopsy sample, where only normal glomeruli are observed, FSGS cannot be differentiated from MCD. The correct diagnosis is crucial to an effective treatment, as MCD is normally responsive to steroid therapy, whereas FSGS is usually resistant. The purpose of our study was to discover and validate novel early urinary biomarkers capable to differentiate between MCD and FSGS. Methods Forty-nine patients biopsy-diagnosed of MCD and primary FSGS were randomly subdivided into a training set (10 MCD, 11 FSGS) and a validation set (14 MCD, 14 FSGS). The urinary proteome of the training set was analyzed by two-dimensional differential gel electrophoresis coupled with mass spectrometry. The proteins identified were quantified by enzyme-linked immunosorbent assay in urine samples from the validation set. Results Urinary concentration of alpha-1 antitrypsin, transferrin, histatin-3 and 39S ribosomal protein L17 was decreased and calretinin was increased in FSGS compared to MCD. These proteins were used to build a decision tree capable to predict patient’s pathology. Conclusions This preliminary study suggests a group of urinary proteins as possible non-invasive biomarkers with potential value in the differential diagnosis of MCD and FSGS. These biomarkers would reduce the number of misdiagnoses, avoiding unnecessary or inadequate treatments. Electronic supplementary material The online version of this article (doi:10.1186/s12882-017-0452-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vanessa Pérez
- Laboratory of Experimental Nephrology, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain. .,Department of Nephrology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Carretera del Canyet s/n, ES-08916, Badalona, Barcelona, Spain.
| | - Dolores López
- Department of Pathology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Ester Boixadera
- Applied Statistics Service, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Meritxell Ibernón
- Department of Nephrology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Carretera del Canyet s/n, ES-08916, Badalona, Barcelona, Spain
| | - Anna Espinal
- Applied Statistics Service, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Josep Bonet
- Department of Nephrology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Carretera del Canyet s/n, ES-08916, Badalona, Barcelona, Spain
| | - Ramón Romero
- Laboratory of Experimental Nephrology, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.,Department of Nephrology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Carretera del Canyet s/n, ES-08916, Badalona, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Badalona, Spain
| |
Collapse
|
19
|
Mandili G, Notarpietro A, Khadjavi A, Allasia M, Battaglia A, Lucatello B, Frea B, Turrini F, Novelli F, Giribaldi G, Destefanis P. Beta-2-glycoprotein-1 and alpha-1-antitrypsin as urinary markers of renal cancer in von Hippel–Lindau patients. Biomarkers 2016; 23:123-130. [DOI: 10.1080/1354750x.2016.1269132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Giorgia Mandili
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Medical School, Turin, Italy
- Center for Experimental Research and Medical Studies (CeRMS), Città della Salute e della Scienza, Ospedale San Giovanni Battista, Turin, Italy
| | - Agata Notarpietro
- Department of Oncology, University of Turin Medical School, Turin, Italy
| | - Amina Khadjavi
- Department of Surgical Sciences, University of Turin Medical School, Turin, Italy
| | - Marco Allasia
- Department of Urology, Azienda Ospedaliera Città della Salute e Della Scienza di Torino – Molinette, Turin, Italy
| | - Antonino Battaglia
- Department of Urology, Azienda Ospedaliera Città della Salute e Della Scienza di Torino – Molinette, Turin, Italy
| | - Barbara Lucatello
- Department of Endocrinology, Diabetology and Metabolism, Azienda Ospedaliera Città della Salute e Della Scienza di Torino – Molinette, Turin, Italy
| | - Bruno Frea
- Department of Urology, Azienda Ospedaliera Città della Salute e Della Scienza di Torino – Molinette, Turin, Italy
| | - Francesco Turrini
- Department of Oncology, University of Turin Medical School, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Medical School, Turin, Italy
- Center for Experimental Research and Medical Studies (CeRMS), Città della Salute e della Scienza, Ospedale San Giovanni Battista, Turin, Italy
- Immunogenetics and Transplantation Biology Unit, Azienda Ospedaliera Città della Salute e della Scienza, Ospedale San Giovanni Battista, Turin, Italy
| | - Giuliana Giribaldi
- Department of Oncology, University of Turin Medical School, Turin, Italy
| | - Paolo Destefanis
- Department of Urology, Azienda Ospedaliera Città della Salute e Della Scienza di Torino – Molinette, Turin, Italy
| |
Collapse
|
20
|
Zhao KW, Murray EJB, Murray SS. HK2 Proximal Tubule Epithelial Cells Synthesize and Secrete Plasma Proteins Predominantly Through the Apical Surface. J Cell Biochem 2016; 118:924-933. [DOI: 10.1002/jcb.25786] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 11/07/2016] [Indexed: 01/19/2023]
Affiliation(s)
- Ke-Wei Zhao
- Geriatric Research; Education and Clinical Center (11E); Veterans Affairs Greater Los Angeles Healthcare System; Sepulveda 91343 California
| | - Elsa J. Brochmann Murray
- Geriatric Research; Education and Clinical Center (11E); Veterans Affairs Greater Los Angeles Healthcare System; Sepulveda 91343 California
- Department of Medicine; University of California; Los Angeles 90095 California
| | - Samuel S. Murray
- Geriatric Research; Education and Clinical Center (11E); Veterans Affairs Greater Los Angeles Healthcare System; Sepulveda 91343 California
- Department of Medicine; University of California; Los Angeles 90095 California
- Interdepartmental Program in Biomedical Engineering; University of California; Los Angeles 90095 California
| |
Collapse
|
21
|
Zager RA, Johnson ACM, Frostad K. An evaluation of the antioxidant protein α1-microglobulin as a renal tubular cytoprotectant. Am J Physiol Renal Physiol 2016; 311:F640-51. [DOI: 10.1152/ajprenal.00264.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/04/2016] [Indexed: 11/22/2022] Open
Abstract
α1-Microglobulin (A1M) is a low-molecular-weight heme-binding antioxidant protein that is readily filtered by the glomerulus and reabsorbed by proximal tubules. Given these properties, recombinant A1M (rA1M) has been proposed as a renal antioxidant and therapeutic agent. However, little direct evidence to support this hypothesis exists. Hence, we have sought “proof of concept” in this regard. Cultured proximal tubule (HK-2) cells or isolated mouse proximal tubule segments were challenged with a variety of prooxidant insults: 1) hemin, 2) myoglobin; 3) “catalytic” iron, 4) H2O2/Fenton reagents, 5) a Ca2+ ionophore, 6) antimycin A, or 7) hypoxia (with or without rA1M treatment). HK-2 injury was gauged by the percent lactate dehydrogenase release and 4,5-(dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide uptake. In vivo protection was sought in rA1M-treated mice subjected to 1) graded myohemoglobinura (2, 4, 8, or 9 ml/kg glycerol injection), 2) purified myoglobinemia/uria, or 3) endotoxemia. In vivo injury was assessed by blood urea nitrogen, creatinine, and the expression of redox-sensitive genes (heme oxygenase-1, neutrophil gelatinase-associated lipocalin, and monocyte chemoattractant protein-1 mRNAs). Although rA1M totally blocked in vitro hemin toxicity, equimolar albumin (another heme binder) or 10% serum induced equal protection. rA1M failed to mitigate any nonhemin forms of either in vitro or in vivo injury. A1M appeared to be rapidly degraded within proximal tubules (by Western blot analysis). Surprisingly, rA1M exerted select injury-promoting effects (increased in vitro catalytic iron/antimycin toxicities and increased in vivo monocyte chemoattractant protein-1/neutrophil gelatinase-associated lipocalin mRNA expression after glycerol or endotoxin injection). We conclude that rA1M has questionable utility as a renal antioxidant/cytoprotective agent, particularly in the presence of larger amounts of competitive free heme (e.g., albumin) binders.
Collapse
Affiliation(s)
- Richard A. Zager
- Fred Hutchinson Cancer Center, Seattle, Washington; and
- University of Washington, Seattle Washington
| | | | | |
Collapse
|
22
|
The neutrophil elastase inhibitor, sivelestat, attenuates sepsis-related kidney injury in rats. Int J Mol Med 2016; 38:767-75. [PMID: 27430552 PMCID: PMC4990314 DOI: 10.3892/ijmm.2016.2665] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/24/2016] [Indexed: 12/18/2022] Open
Abstract
Sepsis-induced acute kidney injury (AKI) represents a major cause of mortality in intensive care units. Sivelestat, a selective inhibitor of neutrophil elastase (NE), can attenuate sepsis-related acute lung injury. However, whether sivelestat can preserve kidney function during sepsis remains unclear. In this study, we thus examined the effects of sivelestat on sepsis-related AKI. Cecal ligation and puncture (CLP) was performed to induce multiple bacterial infection in male Sprague-Dawley rats, and subsequently, 50 or 100 mg/kg sivelestat were administered by intraperitoneal injection immediately after the surgical procedure. In the untreated rats with sepsis, the mean arterial pressure (MAP) and glomerular filtration rate (GFR) were decreased, whereas serum blood urea nitrogen (BUN) and neutrophil gelatinase-associated lipocalin (NGAL) levels were increased. We found that sivelestat promoted the survival of the rats with sepsis, restored the impairment of MAP and GFR, and inhibited the increased BUN and NGAL levels; specifically, the higher dose was more effective. In addition, sivelestat suppressed the CLP-induced macrophage infiltration, the overproduction of pro-inflammatory mediators (tumor necrosis factor-α, interleukin-1β, high-mobility group box 1 and inducible nitric oxide synthase) and serine/threonine kinase (Akt) pathway activation in the rats. Collectively, our data suggest that the inhibition of NE activity with the inhibitor, sivelestat, is beneficial in ameliorating sepsis-related kidney injury.
Collapse
|
23
|
Combined iron sucrose and protoporphyrin treatment protects against ischemic and toxin-mediated acute renal failure. Kidney Int 2016; 90:67-76. [PMID: 27165818 DOI: 10.1016/j.kint.2016.01.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/08/2016] [Accepted: 01/14/2016] [Indexed: 01/07/2023]
Abstract
Tissue preconditioning, whereby various short-term stressors initiate organ resistance to subsequent injury, is well recognized. However, clinical preconditioning of the kidney for protection against acute kidney injury (AKI) has not been established. Here we tested whether a pro-oxidant agent, iron sucrose, combined with a protoporphyrin (Sn protoporphyrin), can induce preconditioning and protect against acute renal failure. Mice were pretreated with iron sucrose, protoporphyrin, cyanocobalamin, iron sucrose and protoporphyrin, or iron sucrose and cyanocobalamin. Eighteen hours later, ischemic, maleate, or glycerol models of AKI were induced, and its severity was assessed the following day (blood urea nitrogen, plasma creatinine concentrations; post-ischemic histology). Agent impact on cytoprotective gene expression (heme oxygenase 1, hepcidin, haptoglobin, hemopexin, α1-antitrypsin, α1-microglobulin, IL-10) was assessed as renal mRNA and protein levels. AKI-associated myocardial injury was gauged by plasma troponin I levels. Combination agent administration upregulated multiple cytoprotective genes and, unlike single agent administration, conferred marked protection against each tested model of acute renal failure. Heme oxygenase was shown to be a marked contributor to this cytoprotective effect. Preconditioning also blunted AKI-induced cardiac troponin release. Thus, iron sucrose and protoporphyrin administration can upregulate diverse cytoprotective genes and protect against acute renal failure. Associated cardiac protection implies potential relevance to both AKI and its associated adverse downstream effects.
Collapse
|
24
|
Zager RA. Marked protection against acute renal and hepatic injury after nitrited myoglobin + tin protoporphyrin administration. Transl Res 2015; 166:485-501. [PMID: 26117289 PMCID: PMC4609608 DOI: 10.1016/j.trsl.2015.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/02/2015] [Indexed: 01/30/2023]
Abstract
The phenomenon known as renal "ischemic preconditioning," whereby an initial ischemic insult induces resistance against subsequent kidney damage, has been well established in the experimental literature. However, a clinically applicable way to safely recapitulate this state has not been defined. We hypothesized that a unique combination of agents (nitrited myoglobin [N-Mgb] + tin protoporphyrin [SnPP]) can achieve these ends safely and synergistically, increasing cytoprotective proteins (eg, heme oxygenase 1 [HO-1], interleukin 10 [IL-10], and haptoglobin) in kidney cells. To test this hypothesis, CD-1 mice received 1 mg of N-Mgb and 1 μmol of SnPP, either alone or in combination. Renal cortical HO-1, haptoglobin, and IL-10 gene expressions (messenger RNA [mRNA], protein levels) were determined 4 and 18 hours later. Cytoresistance to 3 forms of acute kidney injury (AKI; glycerol-induced rhabdomyolysis, maleate nephrotoxicity, and postischemic AKI progression to chronic kidney disease [CKD]) was assessed. To ascertain whether cytoresistance might emerge in extrarenal organs, hepatic HO-1, IL-10, and haptoglobin levels were also measured, and resistance to 25 minutes of hepatic ischemia-reperfusion injury and hepatotoxicity (intraperitoneal glycerol injection) was sought. N-Mgb + SnPP induced additive or synergistic increases in renal HO-1, haptoglobin, and IL-10 mRNA and protein levels (up to 20-fold) without inducing any apparent renal or extrarenal damage. After 18 hours of post-treatment, marked or complete protection against glycerol-induced AKI, maleate-induced AKI, and postischemic AKI progression to CKD had emerged. Combined N-Mgb + SnPP was more protective than either agent alone (assessed in glycerol model). N-Mgb + SnPP also upregulated cytoprotective pathways in liver and induced marked protection against both hepatic ischemia-reperfusion and toxic liver damage. In conclusion, we posit that "preconditioning" with combined administration of N-Mgb + SnPP represents a promising approach for protecting against diverse forms of renal and nonrenal (hepatic) forms of tissue damage.
Collapse
Affiliation(s)
- Richard A Zager
- Fred Hutchinson Cancer Research Center, Seattle, Wash; Department of Medicine, University of Washington, Seattle, Wash.
| |
Collapse
|
25
|
Zhang J, Ahn J, Suh Y, Hwang S, Davis ME, Lee K. Identification of CTLA2A, DEFB29, WFDC15B, SERPINA1F and MUP19 as Novel Tissue-Specific Secretory Factors in Mouse. PLoS One 2015; 10:e0124962. [PMID: 25946105 PMCID: PMC4422522 DOI: 10.1371/journal.pone.0124962] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/19/2015] [Indexed: 01/07/2023] Open
Abstract
Secretory factors in animals play an important role in communication between different cells, tissues and organs. Especially, the secretory factors with specific expression in one tissue may reflect important functions and unique status of that tissue in an organism. In this study, we identified potential tissue-specific secretory factors in the fat, muscle, heart, lung, kidney and liver in the mouse by analyzing microarray data from NCBI’s Gene Expression Omnibus (GEO) public repository and searching and predicting their subcellular location in GeneCards and WoLF PSORT, and then confirmed tissue-specific expression of the genes using semi-quantitative PCR reactions. With this approach, we confirmed 11 lung, 7 liver, 2 heart, 1 heart and muscle, 7 kidney and 2 adipose and liver-specific secretory factors. Among these genes, 1 lung-specific gene - CTLA2A (cytotoxic T lymphocyte-associated protein 2 alpha), 3 kidney-specific genes - SERPINA1F (serpin peptidase inhibitor, Clade A, member 1F), WFDC15B (WAP four-disulfide core domain 15B) and DEFB29 (defensin beta 29) and 1 liver-specific gene - MUP19 (major urinary protein 19) have not been reported as secretory factors. These genes were tagged with hemagglutinin at the 3’end and then transiently transfected to HEK293 cells. Through protein detection in cell lysate and media using Western blotting, we verified secretion of the 5 genes and predicted the potential pathways in which they may participate in the specific tissue through data analysis of GEO profiles. In addition, alternative splicing was detected in transcripts of CTLA2A and SERPINA1F and the corresponding proteins were found not to be secreted in cell culture media. Identification of novel secretory factors through the current study provides a new platform to explore novel secretory factors and a general direction for further study of these genes in the future.
Collapse
Affiliation(s)
- Jibin Zhang
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Jinsoo Ahn
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio, United States of America
| | - Yeunsu Suh
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Seongsoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Gyeonggi, Republic of Korea
| | - Michael E. Davis
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
26
|
Guttman O, Baranovski BM, Schuster R, Kaner Z, Freixo-Lima GS, Bahar N, Kalay N, Mizrahi MI, Brami I, Ochayon DE, Lewis EC. Acute-phase protein α1-anti-trypsin: diverting injurious innate and adaptive immune responses from non-authentic threats. Clin Exp Immunol 2015; 179:161-72. [PMID: 25351931 DOI: 10.1111/cei.12476] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2014] [Indexed: 12/29/2022] Open
Abstract
One would assume that the anti-inflammatory activity of α1-anti-trypsin (AAT) is the result of inhibiting neutrophil enzymes. However, AAT exhibits tolerogenic activities that are difficult to explain by serine-protease inhibition or by reduced inflammatory parameters. Targets outside the serine-protease family have been identified, supporting the notion that elastase inhibition, the only functional factory release criteria for clinical-grade AAT, is over-emphasized. Non-obvious developments in the understanding of AAT biology disqualify it from being a straightforward anti-inflammatory agent: AAT does not block dendritic cell activities, nor does it promote viral and tumour susceptibilities, stunt B lymphocyte responses or render treated patients susceptible to infections; accordingly, outcomes of elevated AAT do not overlap those attained by immunosuppression. Aside from the acute-phase response, AAT rises during the third trimester of pregnancy and also in advanced age. At the molecular level, AAT docks onto cholesterol-rich lipid-rafts and circulating lipid particles, directly binds interleukin (IL)-8, ADAM metallopeptidase domain 17 (ADAM17) and danger-associated molecular pattern (DAMP) molecules, and its activity is lost to smoke, high glucose levels and bacterial proteases, introducing a novel entity - 'relative AAT deficiency'. Unlike immunosuppression, AAT appears to help the immune system to distinguish between desired responses against authentic threats, and unwanted responses fuelled by a positive feedback loop perpetuated by, and at the expense of, inflamed injured innocent bystander cells. With a remarkable clinical safety record, AAT treatment is currently tested in clinical trials for its potential benefit in a variety of categorically distinct pathologies that share at least one common driving force: cell injury.
Collapse
Affiliation(s)
- O Guttman
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology, Beer-Sheva, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zager RA, Johnson ACM, Frostad KB. Acute hepatic ischemic-reperfusion injury induces a renal cortical "stress response," renal "cytoresistance," and an endotoxin hyperresponsive state. Am J Physiol Renal Physiol 2014; 307:F856-68. [PMID: 25080526 DOI: 10.1152/ajprenal.00378.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatic ischemic-reperfusion injury (HIRI) is considered a risk factor for clinical acute kidney injury (AKI). However, HIRI's impact on renal tubular cell homeostasis and subsequent injury responses remain ill-defined. To explore this issue, 30-45 min of partial HIRI was induced in CD-1 mice. Sham-operated or normal mice served as controls. Renal changes and superimposed injury responses (glycerol-induced AKI; endotoxemia) were assessed 2-18 h later. HIRI induced mild azotemia (blood urea nitrogen ∼45 mg/dl) in the absence of renal histologic injury or proteinuria, implying a "prerenal" state. However, marked renal cortical, and isolated proximal tubule, cytoprotective "stress protein" gene induction (neutrophil gelatinase-associated lipocalin, heme oxygenase-1, hemopexin, hepcidin), and increased Toll-like receptor 4 (TLR4) expression resulted (protein/mRNA levels). Ischemia caused release of hepatic heme-based proteins (e.g., cytochrome c) into the circulation. This corresponded with renal cortical oxidant stress (malondialdehyde increases). That hepatic derived factors can evoke redox-sensitive "stress protein" induction was implied by the following: peritoneal dialysate from HIRI mice, soluble hepatic extract, or exogenous cytochrome c each induced the above stress protein(s) either in vivo or in cultured tubule cells. Functional significance of HIRI-induced renal "preconditioning" was indicated by the following: 1) HIRI conferred virtually complete morphologic protection against glycerol-induced AKI (in the absence of hyperbilirubinemia) and 2) HIRI-induced TLR4 upregulation led to a renal endotoxin hyperresponsive state (excess TNF-α/MCP-1 gene induction). In conclusion, HIRI can evoke "renal preconditioning," likely due, in part, to hepatic release of pro-oxidant factors (e.g., cytochrome c) into the systemic circulation. The resulting renal changes can impact subsequent AKI susceptibility and TLR4 pathway-mediated stress.
Collapse
Affiliation(s)
- Richard A Zager
- The Fred Hutchinson Cancer Research Center, Seattle, Washington; and University of Washington, Seattle, Washington
| | - Ali C M Johnson
- The Fred Hutchinson Cancer Research Center, Seattle, Washington; and University of Washington, Seattle, Washington
| | - Kirsten B Frostad
- The Fred Hutchinson Cancer Research Center, Seattle, Washington; and University of Washington, Seattle, Washington
| |
Collapse
|