1
|
Zylberberg AK, Cottle DL, Runting J, Rodrigues G, Tham MS, Jones LK, Cumming HE, Short KM, Zaph C, Smyth IM. Modulating inflammation with interleukin 37 treatment ameliorates murine Autosomal Dominant Polycystic Kidney Disease. Kidney Int 2024; 105:731-743. [PMID: 38158181 DOI: 10.1016/j.kint.2023.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a leading cause of kidney failure and is associated with substantial morbidity and mortality. Interstitial inflammation is attributed to the action of infiltrating macrophages and is a feature thought to aggravate disease progression. Here, we investigated the therapeutic potential of the anti-inflammatory IL37b cytokine as a treatment for ADPKD using genetic mouse models, demonstrating that transgenic expression of human IL37b reduced collecting duct cyst burden in both early and adult-onset ADPKD rodent models. Moreover, injection of recombinant human IL37b could also reduce cyst burden in early onset ADPKD mice, an observation not associated with increased macrophage number at early stages of cyst formation. Interestingly, transgenic IL37b expression also did not alter macrophage numbers in advanced disease. Whole kidney RNA-seq highlighted an IL37b-mediated upregulation of the interferon signaling pathway and single-cell RNA-seq established that these changes originate at least partly from kidney resident macrophages. We further found that blocking type I interferon signaling in mice expressing IL37b resulted in increased cyst number, confirming this as an important pathway by which IL37b exerts its beneficial effects. Thus, our studies show that IL37b promotes interferon signaling in kidney resident macrophages which suppresses cyst initiation, identifying this protein as a potential therapy for ADPKD.
Collapse
Affiliation(s)
- Allara K Zylberberg
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Denny L Cottle
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
| | - Jessica Runting
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Grace Rodrigues
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Ming Shen Tham
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Lynelle K Jones
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Helen E Cumming
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Kieran M Short
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Colby Zaph
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Ian M Smyth
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Gal-9/Tim-3 signaling pathway activation suppresses the generation of Th17 cells and promotes the induction of Foxp3 + regulatory T cells in renal ischemia-reperfusion injury. Mol Immunol 2023; 156:136-147. [PMID: 36921488 DOI: 10.1016/j.molimm.2023.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
CD4+ T cells mediate the pathogenesis of renal ischemia-reperfusion injury (IRI). Emerging research suggests that a Th17/regulatory T cell (Treg) imbalance plays a pivotal role in the development of renal IRI. A recently identified negative checkpoint protein, T cell immunoglobulin domain and mucin domain family 3 (Tim-3), inhibits the immune response by binding to its ligand, galectin-9 (Gal-9). However, the role of the Gal-9/Tim-3 signaling pathway in the regulation of CD4+ T cell subsets in renal IRI remains unclear. In this study, we investigated the effect of the Gal-9/Tim-3 signaling pathway on Th17/Treg subsets in renal IRI using a mouse model. Renal IRI induced the expression of Gal-9 in renal tubular epithelial cells and increased the proportion of Tim-3+ Th17 cells and Tim-3+ forkhead box P3 (Foxp3)+ Treg cells in the ischemia-reperfusion (IR) kidneys. Administration of rAAV9-Gal-9 suppressed kidney inflammation, reduced the mortality of mice with renal IRI, increased Foxp3+ Treg cells, and reduced Th17 cells. In contrast, the blockade of Tim-3 in vivo using an anti-Tim-3 monoclonal antibody aggravated renal inflammation, decreased Foxp3+ Treg cells, and promoted Th17 cells. Thus, Gal-9/Tim-3 signaling pathway activation may protect against renal IRI by inhibiting Th17 cell production and inducing Foxp3+ Treg cell expansion. Our study suggests that the Gal-9/Tim-3 signaling pathway may be targeted by immunotherapy in renal IRI.
Collapse
|
3
|
Pang H, Kumar S, Ely EW, Gezalian MM, Lahiri S. Acute kidney injury-associated delirium: a review of clinical and pathophysiological mechanisms. Crit Care 2022; 26:258. [PMID: 36030220 PMCID: PMC9420275 DOI: 10.1186/s13054-022-04131-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/20/2022] [Indexed: 11/10/2022] Open
Abstract
Acute kidney injury is a known clinical risk factor for delirium, an acute cognitive dysfunction that is commonly encountered in the critically ill population. In this comprehensive review of clinical and basic research studies, we detail the epidemiology, clinical implications, pathogenesis, and management strategies of patients with acute kidney injury-associated delirium. Specifically addressed are the pathological roles of endogenous toxin or drug accumulation, acute kidney injury-mediated neuroinflammation, and acute kidney injury-associated volume overload as discrete potential biological mechanisms of the condition. The optimization of clinical contributors and normalization of renal function are reviewed as pragmatic management strategies in addition to potential and emerging therapeutic approaches.
Collapse
|
4
|
Odum JD, Standage S, Alder M, Zingarelli B, Devarajan P, Wong HR. Candidate Biomarkers for Sepsis-Associated Acute Kidney Injury Mechanistic Studies. Shock 2022; 57:687-693. [PMID: 35234208 PMCID: PMC9117431 DOI: 10.1097/shk.0000000000001916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Sepsis-associated acute kidney injury (SA-AKI) is a frequent complication of sepsis, yet the pathophysiologic mechanisms of SA-AKI are incompletely understood. PERSEVERE is a clinically validated serum biomarker panel with high sensitivity in predicting mortality from sepsis, and recent evidence suggests it can also predict severe, persistent SA-AKI at day 3 of hospitalization among septic children. We developed a murine model of PERSEVERE (mPERSEVERE) to further interrogate the sepsis-related biological underpinnings of SA-AKI using candidate biomarkers within mPERSEVERE. METHODS Eight-week-old C57BL/6 male mice underwent induction of sepsis by cecal ligation and puncture (CLP). mPERSEVERE biomarkers were collected at 8-hours and kidneys were harvested at 24-hours post-CLP Classification and regression tree analysis (CART) was used to generate a SA-AKI predictive model. Kidney gene expression levels of candidate biomarkers were quantified using real time polymerase chain reaction. RESULTS Thirty- five mice underwent CLP Among mice identified by mPERSEVERE as high-risk for mortality, 70% developed SA-AKI at 24-hours compared to 22% of low-risk mice. CART analysis identified two mPERSEVERE biomarkers-C-C motif chemokine ligand 3 (CCL3) and keratinocyte-derived chemokine (KC)-as most predictive for SA-AKI with an area under the receiver operating curve of 0.90. In mice that developed SA-AKI, renal expression of KC was significantly increased compared to mice without SA-AKI (p = 0.013), whereas no difference was seen in renal expression of CCL3 in mice with SA-AKI vs. no SA-AKI. KC and CCL3 localized to renal tubule epithelial cells as opposed to infiltrating immune cells by immunohistochemistry. CONCLUSIONS The combination of plasma CCL3+KC can predict SA-AKI development in mice at 24-hours following CLP Of these two biomarkers, only renal expression of KC is increased in mice with SA-AKI. Further studies are required to determine if KC directly contributes to the underlying pathobiology of SA-AKI.
Collapse
Affiliation(s)
- James D Odum
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Steve Standage
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Matthew Alder
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Basilia Zingarelli
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Prasad Devarajan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hector R Wong
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
5
|
Satta E, Alfarone C, De Maio A, Gentile S, Romano C, Polverino M, Polverino F. Kidney and lung in pathology: mechanisms and clinical implications. Multidiscip Respir Med 2022; 17:819. [PMID: 35127080 PMCID: PMC8791019 DOI: 10.4081/mrm.2022.819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/04/2021] [Indexed: 11/23/2022] Open
Abstract
There is a close, physiological, relationship between kidney and lung that begin in the fetal age, and is aimed to keep homeostatic balance in the body. From a pathological point of view, the kidneys could be damaged by inflammatory mediators or by immune-mediated factors linked to a primary lung disease or, conversely, it could be the kidney disease that causes lung damage. Non-immunological mechanisms are frequently involved in renal and pulmonary diseases, as observed in chronic conditions. This crosstalk have clinical and therapeutic consequences. This review aims to describe the pulmonary-renal link in physiology and in pathological conditions.
Collapse
|
6
|
Lu XH, Zhang J, Xiong Q. Suppressive effect erythropoietin on oxidative stress by targeting AMPK/Nox4/ROS pathway in renal ischemia reperfusion injury. Transpl Immunol 2022; 72:101537. [PMID: 35031454 DOI: 10.1016/j.trim.2022.101537] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/09/2022] [Accepted: 01/09/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To explore the effect of erythropoietin (EPO) on the AMP-activated protein kinase (AMPK)/nicotinamide adenine dinucleotide phosphatase oxidase 4 (NOX4) signaling pathway during renal ischemia reperfusion injury (RIRI) in rats. METHODS A rat model of RIRI was established by clamping the left renal pedicle and removing the right kidney. The rats in the sham group did not have their left renal pedicle clamped. Rats with a model of RIRI were randomly divided into RIRI alone (control), erythropoietin treatment (EPO/RIRI), and Compound C treatment (CPC/RIRI) groups. Hematoxylin-eosin (H&E) staining was used to examine pathological kidney damage. Serum creatinine and urea nitrogen levels were measured to evaluate renal function. Western blotting was performed to detect the expression levels of phosphorylated p-AMPK and total AMPK protein in the kidneys. RT-PCR was used to evaluate the mRNA levels of Nox4 and p22 in the kidneys. Oxidative stress-related indices (ROS, CAT, GSH, SOD, and MDA) were also measured. RESULTS EPO treatment improved kidney function by preventing kidney damage induced by the RIRI model. Preventing ischemia/reperfusion injury in the RIRI model was correlated with an increased p-AMPK/AMPK ratio and elevated activity of CAT, GSH, and SOD, which ameliorated the expression of NOX4, p22, ROS, and MDA. Moreover, treatment with CPC (an AMPK inhibitor) reduced the effects of EPO in the RIRI model. CONCLUSION EPO treatment protected rats against RIRI in the RIRI model by alleviating oxidative stress by triggering the AMPK/NOX4/ROS pathway.
Collapse
Affiliation(s)
- Xiang-Heng Lu
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jiong Zhang
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Disease, Chengdu 610072, China
| | - Qin Xiong
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
7
|
Agrawal YO, Mahajan UB, Mahajan HS, Ojha S. Methotrexate-Loaded Nanostructured Lipid Carrier Gel Alleviates Imiquimod-Induced Psoriasis by Moderating Inflammation: Formulation, Optimization, Characterization, In-Vitro and In-Vivo Studies. Int J Nanomedicine 2020; 15:4763-4778. [PMID: 32753865 PMCID: PMC7354956 DOI: 10.2147/ijn.s247007] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Methotrexate exhibits poor cutaneous bioavailability and systemic side effects on topical administration, so there is an unmet need for a novel carrier and its optimized therapy. Methotrexate-loaded nanostructured lipid carriers (MTXNLCs) were formulated and characterized to determine in vitro drug release and evaluate the role of MTXNLC gel in the topical treatment of psoriasis. METHODS A solvent diffusion technique was employed to prepare MTXNLCs, which was optimized using 32 full factorial designs. The mean diameter and surface morphology of MTXNLCs was evaluated. The crystallinity of lyophilized MTXNLCs was characterized by differential scanning calorimetry (DSC) and powder X-ray diffraction (XRD). MTXNLCs were integrated in 1% w/w Carbopol 934 P gel base, and in vitro skin deposition studies in human cadaver skin (HCS) were carried out. RESULTS The optimized MTXNLCs were rod-shaped, with an average particle size of 253 ± 8.65 nm, a zeta potential of -26.4±0.86 mV, and EE of 54.00±1.49%. DSC and XRD data confirmed the formation of NLCs. Significantly higher deposition of MTX was found in HCS from MTXNLC gel (71.52 ±1.13%) as compared to MTX plain gel (38.48±0.96%). In vivo studies demonstrated significant improvement in therapeutic response and reduction in local side effects with MTXNLCs-loaded gel in the topical treatment of psoriasis. Anti-psoriatic efficacy of MTXNLCs 100 ug/cm2 compared with plain MTX gel was evaluated using imiquimod (IMQ)-induced psoriasis in BALB/c mice. The topical application of MTXNLCs to the mouse ear resulted in a significant reduction of psoriatic area and severity index, oxidative stress, inflammatory cytokines like TNF-α, IL-1β, and IL-6 and IMQ-induced histopathological alterations in mouse ear samples. CONCLUSION Developed formulation of MTXNLC gel demonstrated better anti-psoriatic activity and also displayed prolonged and sustained release effect, which shows that it can be a promising alternative to existing MTX formulation for the treatment of psoriasis.
Collapse
Affiliation(s)
- Yogeeta O Agrawal
- Department of Pharmaceutics and Quality Assurance, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra425405, India
| | - Umesh B Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra425405, India
| | - Hitendra S Mahajan
- Department of Pharmaceutics and Quality Assurance, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra425405, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
8
|
Remote organ failure in acute kidney injury. J Formos Med Assoc 2019; 118:859-866. [DOI: 10.1016/j.jfma.2018.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/09/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023] Open
|
9
|
Dingwell LS, Shikatani EA, Besla R, Levy AS, Dinh DD, Momen A, Zhang H, Afroze T, Chen MB, Chiu F, Simmons CA, Billia F, Gommerman JL, John R, Heximer S, Scholey JW, Bolz SS, Robbins CS, Husain M. B-Cell Deficiency Lowers Blood Pressure in Mice. Hypertension 2019; 73:561-570. [DOI: 10.1161/hypertensionaha.118.11828] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Luke S. Dingwell
- From the Toronto General Hospital Research Institute, University Health Network, Canada (L.S.D., E.A.S., A.M., T.A., F.B., M.H.)
- Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre (L.S.D., E.A.S., C.S.R., M.H.), University of Toronto, Canada
- Department of the Institute of Medical Science (L.S.D., M.H.), University of Toronto, Canada
| | - Eric A. Shikatani
- From the Toronto General Hospital Research Institute, University Health Network, Canada (L.S.D., E.A.S., A.M., T.A., F.B., M.H.)
- Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre (L.S.D., E.A.S., C.S.R., M.H.), University of Toronto, Canada
- Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., F.C., R.J., C.S.R., M.H.), University of Toronto, Canada
| | - Rickvinder Besla
- Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., F.C., R.J., C.S.R., M.H.), University of Toronto, Canada
| | - Andrew S. Levy
- Department of Physiology (A.S.L., D.D.D., H.Z., S.H., J.W.S., S.-S.B., M.H.), University of Toronto, Canada
| | - Danny D. Dinh
- Department of Physiology (A.S.L., D.D.D., H.Z., S.H., J.W.S., S.-S.B., M.H.), University of Toronto, Canada
| | - Abdul Momen
- From the Toronto General Hospital Research Institute, University Health Network, Canada (L.S.D., E.A.S., A.M., T.A., F.B., M.H.)
| | - Hangjun Zhang
- Department of Physiology (A.S.L., D.D.D., H.Z., S.H., J.W.S., S.-S.B., M.H.), University of Toronto, Canada
| | - Talat Afroze
- From the Toronto General Hospital Research Institute, University Health Network, Canada (L.S.D., E.A.S., A.M., T.A., F.B., M.H.)
| | - Michelle B. Chen
- Department of Mechanical and Industrial Engineering (M.B.C., C.A.S.), University of Toronto, Canada
| | - Felix Chiu
- Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., F.C., R.J., C.S.R., M.H.), University of Toronto, Canada
| | - Craig A. Simmons
- Department of Mechanical and Industrial Engineering (M.B.C., C.A.S.), University of Toronto, Canada
| | - Filio Billia
- From the Toronto General Hospital Research Institute, University Health Network, Canada (L.S.D., E.A.S., A.M., T.A., F.B., M.H.)
| | | | - Rohan John
- Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., F.C., R.J., C.S.R., M.H.), University of Toronto, Canada
| | - Scott Heximer
- Department of Physiology (A.S.L., D.D.D., H.Z., S.H., J.W.S., S.-S.B., M.H.), University of Toronto, Canada
| | - James W. Scholey
- Department of Mechanical and Industrial Engineering (M.B.C., C.A.S.), University of Toronto, Canada
| | - Steffen-Sebastian Bolz
- Department of Mechanical and Industrial Engineering (M.B.C., C.A.S.), University of Toronto, Canada
| | - Clinton S. Robbins
- Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre (L.S.D., E.A.S., C.S.R., M.H.), University of Toronto, Canada
- Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., F.C., R.J., C.S.R., M.H.), University of Toronto, Canada
- Department of Immunology (J.L.G., C.S.R.), University of Toronto, Canada
| | - Mansoor Husain
- From the Toronto General Hospital Research Institute, University Health Network, Canada (L.S.D., E.A.S., A.M., T.A., F.B., M.H.)
- Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre (L.S.D., E.A.S., C.S.R., M.H.), University of Toronto, Canada
- Department of the Institute of Medical Science (L.S.D., M.H.), University of Toronto, Canada
- Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., F.C., R.J., C.S.R., M.H.), University of Toronto, Canada
- Department of Physiology (A.S.L., D.D.D., H.Z., S.H., J.W.S., S.-S.B., M.H.), University of Toronto, Canada
| |
Collapse
|
10
|
Su M, Hu X, Lin J, Zhang L, Sun W, Zhang J, Tian Y, Qiu W. Identification of Candidate Genes Involved in Renal Ischemia/Reperfusion Injury. DNA Cell Biol 2019; 38:256-262. [PMID: 30668132 PMCID: PMC6434600 DOI: 10.1089/dna.2018.4551] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Renal ischemia/reperfusion injury (IRI) is a main risk factor for the occurrence of delayed graft function or primary graft nonfunction of kidney transplantation. However, it lacks ideal molecular markers for indicating IRI in kidney transplantation. The present study is to explore novel candidate genes involved in renal IRI. Experimental renal IRI mouse models were constructed, and the differentially expressed genes were screened using a microarray assay. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed. The expression of genes was detected using real-time qPCR assay. Western blotting and immunohistochemistry staining assays were performed for protein determination. We identified that renal IRI induced the upregulation of SPRR2F, SPRR1A, MMP-10, and long noncoding RNA (lncRNA) Malat1 in kidney tissues for 479.3-, 4.98-, 238.1-, and 3.79-fold, respectively. The expression of miR-139-5p in kidney tissues of IRI-treated mice was decreased to 40.4% compared with the sham-operated mice. These genes are associated with keratinocyte differentiation, regeneration and repair of kidney tissues, extracellular matrix degradation and remodeling, inflammation, and cell proliferation in renal IRI. Identification of novel biomarkers involved in renal IRI may provide evidences for the diagnosis and treatment of renal IRI.
Collapse
Affiliation(s)
- Ming Su
- 1 Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xinyi Hu
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jun Lin
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lei Zhang
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wen Sun
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jian Zhang
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ye Tian
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Qiu
- 2 Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
11
|
Urbschat A, Baer P, Zacharowski K, Sprunck V, Scheller B, Raimann F, Maier TJ, Hegele A, Hofmann R, Mersmann J. Systemic TLR2 Antibody Application in Renal Ischaemia and Reperfusion Injury Decreases AKT Phosphorylation and Increases Apoptosis in the Mouse Kidney. Basic Clin Pharmacol Toxicol 2017; 122:223-232. [PMID: 28857508 DOI: 10.1111/bcpt.12896] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/21/2017] [Indexed: 12/27/2022]
Abstract
Acute kidney injury remains an important cause of renal dysfunction. In this context, Toll-like receptors have been demonstrated to play a critical role in the induction of innate and inflammatory responses. Among these, Toll-like receptor 2 (TLR2) is constitutively expressed in tubular epithelial cells (TECs) of the kidney and is also known to mediate ischaemia reperfusion (IR) injury. Adult male C57BL/6JRj mice were randomized into seven groups (n = 8): a non-operative control group (CTRL) and six interventional groups in which mice were subjected to a 30 min. bilateral renal ischaemia. Immediately before reperfusion, mice were treated either with saline or with TLR2 antibody (clone T2.5) and harvested after ischaemia and reperfusion for 3, 24 and 48 hr. Analysed kidney homogenates of TLR2 antibody-treated mice displayed significantly decreased levels of TLR2 protein after 3 hr of IR compared to saline-treated mice. Accordingly, the degree of AKT phosphorylation was significantly decreased after 3 hr of IR compared to saline-treated animals. TUNEL staining revealed significantly higher apoptosis rates in TLR2 antibody-treated animals compared to saline-treated mice after 3 and 24 hr of IR. Further, a positive correlation between TLR2 protein expression and phosphorylation of AKT as well as a negative correlation with the number of TUNEL-positive cells could be observed. Inhibition of TLR2 and its signalling pathway by a single application of TLR2 antibody results in reduced phosphorylation of AKT and consecutively increased apoptosis.
Collapse
Affiliation(s)
- Anja Urbschat
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Clinic of Urology and Pediatric Urology, Medical School, Philipps-University Marburg, Marburg, Germany
| | - Patrick Baer
- Clinic of Internal Medicine III, Division of Nephrology, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Kai Zacharowski
- Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Vera Sprunck
- Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Bertram Scheller
- Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Florian Raimann
- Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Thorsten Jürgen Maier
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Axel Hegele
- Clinic of Urology and Pediatric Urology, Medical School, Philipps-University Marburg, Marburg, Germany
| | - Rainer Hofmann
- Clinic of Urology and Pediatric Urology, Medical School, Philipps-University Marburg, Marburg, Germany
| | - Jan Mersmann
- Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
12
|
Identification of Novel Inflammatory Cytokines and Contribution of Keratinocyte-Derived Chemokine to Inflammation in Response to Vibrio vulnificus Infection in Mice. Inflammation 2016; 38:1864-73. [PMID: 25862020 DOI: 10.1007/s10753-015-0166-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Currently, only tumor necrosis factor alpha (TNF-α) and interleukin family cytokines have been found to be elicited in Vibrio vulnificus (V. vulnificus)-infected animal models and humans. However, multiple other cytokines are also involved in the immune and inflammatory responses to foreign microorganism infection. Antibody array technology, unlike traditional enzyme-linked immunosorbent assay (ELISA), is able to detect multiple cytokines at one time. Therefore, in this study, we examined the proinflammatory cytokine profile in the serum and liver homogenate samples of bacterial-infected mice using antibody array technology. We identified nine novel cytokines in response to V. vulnificus infection in mice. We found that keratinocyte-derived chemokine (KC) was the most elevated cytokine and demonstrated that KC played a very important role in the V. vulnificus infection-elicited inflammatory response in mice, as evidenced by the fact that the blocking of KC by anti-KC antibody reduced hepatic injury in vivo and that KC induced by V. vulnificus infection in AML-12 cells chemoattracted neutrophils. Our findings implicate that KC may serve as a novel diagnostic biomarker and a possible therapeutic target for V. vulnificus infection.
Collapse
|
13
|
Li Z, Deng X, Kang Z, Wang Y, Xia T, Ding N, Yin Y. Elevation of miR-21, through targeting MKK3, may be involved in ischemia pretreatment protection from ischemia-reperfusion induced kidney injury. J Nephrol 2016; 29:27-36. [PMID: 26149640 DOI: 10.1007/s40620-015-0217-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/19/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Ischemia-reperfusion (IR) causes acute kidney injury (AKI), and ischemia pretreatment may exert protection. Mitogen-activated protein kinase kinase 3 (MKK3), which is involved in the signal transduction pathway in IR-induced injury, is a potential target of miR-21. We aimed to verify the targeting regulation of miR-21 on MKK3 and to explore the effects of miR-21-mediated MKK3 expression changes in AKI. METHODS Vectors containing the MKK3 3'UTR and mutated MKK3-3U-M were constructed and co-transfected with nonsense miR, miR-21-5p mimics or inhibitor in HEK293 cells. Gene expressions were detected by dual luciferase reporter assay. The effects of miR-21 on mRNA and protein of MKK3 were investigated in HK-2 cells. Male C57BL/6J mice were treated with ischemic preconditioning (IPC) and IR. Kidney functions were assessed through monitoring serum creatinine (Scr) and blood urea nitrogen (BUN). Pathological changes were observed and scored with histological samples of kidney. Expression levels of miR-21, MKK3, interleukin (IL)-6, tumor necrosis factor (TNF)-α before and after IPC and IR were examined by real-time polymerase chain reaction and/or immunohistochemistry. RESULTS miR-21 regulated the expression of MKK3 via 3'UTR. Following IR, MKK3, IL-6 and TNF-α levels were increased. Scr, BUN and pathological injuries were aggravated, and miR-21 expression was increased. IPC increased miR-21 levels ahead of IR and inhibited the increases in MKK3, IL-6 and TNF-α levels and the aggravation of Scr, BUN and pathological injuries. CONCLUSIONS miR-21 targets MKK3 in vivo and in vitro, inhibiting the downstream factors IL-6 and TNF-α. Therefore, miR-21 might be involved in protection of IPC against IR of the kidney.
Collapse
Affiliation(s)
- Zhihui Li
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China.
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China.
| | - Xu Deng
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Zhijuan Kang
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Ying Wang
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Tuanhong Xia
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Niu Ding
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Yan Yin
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| |
Collapse
|
14
|
Stimulation of Dopamine D3 Receptor Attenuates Renal Ischemia-Reperfusion Injury via Increased Linkage With Gα12. Transplantation 2016; 99:2274-84. [PMID: 25989500 DOI: 10.1097/tp.0000000000000762] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Renal ischemia-reperfusion (I/R) injury causes renal tubular necrosis, apoptosis, and inflammation leading to acute renal dysfunction. Recent studies have revealed that deletion of Gα12 mitigates the renal damage due to I/R injury. Our previous study showed that activation of dopamine D3 receptor (D3R) increased its linkage with Gα12, and hampered Gα12-mediated stimulation of renal sodium transport. In the present study, we used an in-vivo rat model and an in vitro study of the renal epithelial cell line (NRK52E) to investigate whether or not an increased linkage between D3R and Gα12 contributes to the protective effect of D3R on renal I/R injury. METHODS For in vivo studies, I/R injury was induced in a rat renal unilateral clamping model. For in vitro studies, hypoxia/reoxygenation and cold storage/rewarming injuries were performed in NRK52E cells. PD128907, a D3R agonist, or vehicle, was administered 15 minutes before clamping (or hypoxia) in both the in vivo or in vitro studies. RESULTS In the rat renal unilateral clamping model, pretreatment with PD128907 (0.2 mg/kg, intravenous) protected against renal I/R injury and increased survival rate during a long-term follow-up after 7 days. A decrease in the generation of reactive oxygen species, apoptosis, and inflammation may be involved in the D3R-mediated protection because pretreatment with PD128907 increased renal glutathione and superoxide dismutase levels and decreased malondialdehyde levels in the I/R group. The increase in cytokines (TNF-α, IL-1β, and IL-10) and myeloperoxidase in I/R injured kidney was also prevented with a simultaneous decrease in the apoptosis of the epithelial cells and expression of apoptosis biomarkers in kidney harvested 1 day after I/R injury. The increase in the coimmunoprecipitation between D3R and Gα12 with D3R stimulation paralleled the observed renal protection from I/R injury. Moreover, in vitro studies showed that transient overexpression of Gα12 in the NRK52E cells attenuated the protective effect of PD128907 on hypoxia/reoxygenation injury. The protective effect of PD128907 might be of significance to renal transplantation because cold storage/rewarming induced injury increased lactate dehydrogenase release and decreased cell viability in NRK52E cells. Conversely, in the presence of PD128907, the increased lactate dehydrogenase release and decreased cell viability were reversed. CONCLUSIONS These results suggest that activation of D3R, by decreasing Gα12-induced renal damage, may exert a protective effect from I/R injury.
Collapse
|
15
|
Bersani-Amado LE, Dantas JA, Damião MJ, Rocha BA, Besson JCF, Bastos RL, Silva LN, Bersani-Amado CA, Cuman RKN. Involvement of cytokines in the modulation and progression of renal fibrosis induced by unilateral ureteral obstruction in C57BL/6 mice: effects of thalidomide and dexamethasone. Fundam Clin Pharmacol 2015; 30:35-46. [PMID: 26501392 DOI: 10.1111/fcp.12162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/25/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
This study investigated the kinetics of cytokines that are involved in the development of interstitial fibrosis in mice that were subjected to UUO, the interstitial type I and III collagen deposition, and the effects of Thalido and Dexa treatment on these parameters. Inbred C57BL/6 mice were divided into the groups: Normal (not submitted surgery), Sham (sham surgery), Control (UUO treated with 0.5% carboxymethyl cellulose), Thalido (UUO treated with 5 mg/kg thalidomide), and Dexa (UUO treated with 1 mg/kg dexamethasone). The treatments began the day before surgery and were administered once daily by gavage for 1, 7, or 14 days. At the end of each treatment period, blood samples were collected for the determination of creatinine, urea, cytokines. The Control group exhibited a increase in creatinine concentration compared with the Normal and Sham groups within the first 24 h after UUO, which remained high until days 7 and 14. The urea concentration was higher on days 7 and 14 in the Control group compared with the Sham group. In the Thalido and Dexa groups, a reduction of serum creatinine concentration was seen on day 14. Treatment with Dexa reduced the serum concentration of urea on day 7. The serum concentrations of cytokines (TNF-α, IL-1β, IL-6, IL-10 and IL-17) and chemokines (KC, MIG, bFGF) increased in UUO mice at all of the sampling times. The Dexa and Thalido groups exhibited alterations in the concentrations of these cytokines, suggesting the involvement of anti-inflammatory and immunomodulatory mechanisms that may have modified the fibrosis framework.
Collapse
Affiliation(s)
| | - Jaílson Araujo Dantas
- Department of Pharmacology and Therapeutic-State University of Maringá, Avenida Colombo, 5790, Maringá, Paraná, Brazil
| | - Marcio José Damião
- Department of Pharmacology and Therapeutic-State University of Maringá, Avenida Colombo, 5790, Maringá, Paraná, Brazil
| | - Bruno Ambrósio Rocha
- Department of Pharmacology and Therapeutic-State University of Maringá, Avenida Colombo, 5790, Maringá, Paraná, Brazil
| | - Jean Carlos Fernando Besson
- Department of Morphological Sciences-State University of Maringá, Avenida Colombo, 5790, Maringá, Paraná, Brazil
| | - Rafael Lucena Bastos
- Fellowship (Medicine), State University of Maringá, Avenida Colombo, 5790, Maringá, Paraná, Brazil
| | - Letícia Nicoletti Silva
- Fellowship (medicine) Evangelical Faculty of Paraná, Rua Padre Anchieta, 2770, Curitiba, Paraná, Brazil
| | | | - Roberto Kenji Nakamura Cuman
- Department of Pharmacology and Therapeutic-State University of Maringá, Avenida Colombo, 5790, Maringá, Paraná, Brazil
| |
Collapse
|
16
|
An experimental model-based exploration of cytokines in ablative radiation-induced lung injury in vivo and in vitro. Lung 2015; 193:409-19. [PMID: 25749666 DOI: 10.1007/s00408-015-9705-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/26/2015] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Stereotactic ablative radiotherapy is a newly emerging radiotherapy treatment method that, compared with conventionally fractionated radiation therapy (CFRT), allows an ablative dose of radiation to be delivered to a confined area around a tumor. The aim of the present study was to investigate the changes of various cytokines that may be involved in ablative radiation-induced lung injury in vitro and in vivo. METHODS In the in vivo study, ablative-dose radiation was delivered to a small volume of the left lung of C3H/HeJCr mice using a small-animal irradiator. The levels of 24 cytokines in the peripheral blood were tested at several time points after irradiation. For the in vitro study, three mouse cell types (type II pneumocytes, alveolar macrophages, and fibroblasts) known to play important roles in radiation-induced pneumonitis and lung fibrosis were analyzed using a co-culture system. RESULTS In the in vivo study, we found obvious patterns of serum cytokine changes depending on the volume of tissue irradiated (2-mm vs. 3.5-mm collimator). Only the levels of 3 cytokines increased with the 2-mm collimator at the acute phase (1-2 weeks after irradiation), while the majority of cytokines were elevated with the 3.5-mm collimator. In the in vitro co-culture system, after the cells were given an ablative dose of irradiation, the levels of five cytokines (GM-CSF, G-CSF, IL-6, MCP-1, and KC) increased significantly in a dose-dependent manner. CONCLUSIONS The cytokine levels in our radiation-induced lung injury model showed specific changes, both in vivo and in vitro. These results imply that biological studies related to ablative-dose small-volume irradiation should be investigated using the corresponding experimental models rather than on those simulating large-volume CFRT.
Collapse
|
17
|
Huang JX, Blaskovich MA, Cooper MA. Cell- and biomarker-based assays for predicting nephrotoxicity. Expert Opin Drug Metab Toxicol 2014; 10:1621-35. [DOI: 10.1517/17425255.2014.967681] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Chou AH, Lee CM, Chen CY, Liou JT, Liu FC, Chen YL, Day YJ. Hippocampal transcriptional dysregulation after renal ischemia and reperfusion. Brain Res 2014; 1582:197-210. [DOI: 10.1016/j.brainres.2014.07.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/12/2014] [Accepted: 07/17/2014] [Indexed: 11/26/2022]
|
19
|
Liang CJ, Shen WC, Chang FB, Wu VC, Wang SH, Young GH, Tsai JS, Tseng YC, Peng YS, Chen YL. Endothelial Progenitor Cells Derived From Wharton's Jelly of Human Umbilical Cord Attenuate Ischemic Acute Kidney Injury by Increasing Vascularization and Decreasing Apoptosis, Inflammation, and Fibrosis. Cell Transplant 2014; 24:1363-77. [PMID: 24819279 DOI: 10.3727/096368914x681720] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury to the kidney, a major cause of acute renal failure in humans, is associated with a high mortality, and the development of a new therapeutic strategy is therefore highly desirable. In this study, we examined the therapeutic potential of implantation of endothelial progenitor cells (EPCs) isolated from Wharton's jelly of human umbilical cords in the treatment of renal I/R injury in mice. To visualize the localization of the transplanted EPCs, the cells were labeled with Q-tracker before injection into the renal capsule. Mice with renal I/R injury showed a significant increase in blood urea nitrogen and creatinine levels, and these effects were decreased by EPC transplantation. The kidney injury score in the mice with I/R injury was also significantly decreased by EPC transplantation. EPC transplantation increased the microvascular density, and some of the EPCs surrounded and were incorporated into microvessels. In addition, EPC transplantation inhibited the I/R-induced cell apoptosis of endothelial, glomerular, and renal tubular cells, as demonstrated by TUNEL staining, and significantly reduced reactive oxygen species production and the expression of the inflammatory chemokines macrophage inflammatory protein-2 and keratinocyte-derived cytokine, as shown by immunostaining and ELISA. Moreover, EPC transplantation reduced I/R-induced fibrosis, as demonstrated by immunostaining for S100A4, a fibroblast marker, and by Jones silver staining. To our knowledge, this is the first report that transplantation of EPCs from Wharton's jelly of human umbilical cords might provide a novel therapy for ischemic acute kidney injury by promoting angiogenesis and inhibiting apoptosis, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Chan-Jung Liang
- Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Thurman JM, Serkova NJ. Nanosized contrast agents to noninvasively detect kidney inflammation by magnetic resonance imaging. Adv Chronic Kidney Dis 2013; 20:488-99. [PMID: 24206601 DOI: 10.1053/j.ackd.2013.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 06/02/2013] [Accepted: 06/03/2013] [Indexed: 12/15/2022]
Abstract
Several molecular imaging methods have been developed that use nanosized contrast agents to detect markers of inflammation within tissues. Kidney inflammation contributes to disease progression in a wide range of autoimmune and inflammatory diseases, and a biopsy is currently the only method of definitively diagnosing active kidney inflammation. However, the development of new molecular imaging methods that use contrast agents capable of detecting particular immune cells or protein biomarkers will allow clinicians to evaluate inflammation throughout the kidneys and to assess a patient's response to immunomodulatory drugs. These imaging tools will improve our ability to validate new therapies and to optimize the treatment of individual patients with existing therapies. This review describes the clinical need for new methods of monitoring kidney inflammation and recent advances in the development of nanosized contrast agents for the detection of inflammatory markers of kidney disease.
Collapse
|
21
|
Kim MG, Koo TY, Yan JJ, Lee E, Han KH, Jeong JC, Ro H, Kim BS, Jo SK, Oh KH, Surh CD, Ahn C, Yang J. IL-2/anti-IL-2 complex attenuates renal ischemia-reperfusion injury through expansion of regulatory T cells. J Am Soc Nephrol 2013; 24:1529-36. [PMID: 23833258 DOI: 10.1681/asn.2012080784] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Regulatory T cells (Tregs) can suppress immunologic damage in renal ischemia-reperfusion injury (IRI), but the isolation and ex vivo expansion of these cells for clinical application remains challenging. Here, we investigated whether the IL-2/anti-IL-2 complex (IL-2C), a mediator of Treg expansion, can attenuate renal IRI in mice. IL-2C administered before bilateral renal IRI induced Treg expansion in both spleen and kidney, improved renal function, and attenuated histologic renal injury and apoptosis after IRI. Furthermore, IL-2C administration reduced the expression of inflammatory cytokines and attenuated the infiltration of neutrophils and macrophages in renal tissue. Depletion of Tregs with anti-CD25 antibodies abrogated the beneficial effects of IL-2C. However, IL-2C-mediated renal protection was not dependent on either IL-10 or TGF-β. Notably, IL-2C administered after IRI also enhanced Treg expansion in spleen and kidney, increased tubular cell proliferation, improved renal function, and reduced renal fibrosis. In conclusion, these results indicate that IL-2C-induced Treg expansion attenuates acute renal damage and improves renal recovery in vivo, suggesting that IL-2C may be a therapeutic strategy for renal IRI.
Collapse
Affiliation(s)
- Myung-Gyu Kim
- Transplantation Center, Seoul National University Hospital, Seoul
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
White RL, Nash G, Kavanagh DPJ, Savage COS, Kalia N. Modulating the Adhesion of Haematopoietic Stem Cells with Chemokines to Enhance Their Recruitment to the Ischaemically Injured Murine Kidney. PLoS One 2013; 8:e66489. [PMID: 23840488 PMCID: PMC3686749 DOI: 10.1371/journal.pone.0066489] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/07/2013] [Indexed: 02/06/2023] Open
Abstract
Introduction Renal disease affects over 500 million people worldwide and is set to increase as treatment options are predominately supportive. Evidence suggests that exogenous haematopoietic stem cells (HSCs) can be of benefit but due to the rarity and poor homing of these cells, benefits are either minor or transitory. Mechanisms governing HSC recruitment to injured renal microcirculation are poorly understood; therefore this study determined (i) the adhesion molecules responsible for HSC recruitment to the injured kidney, (ii) if cytokine HSC pre-treatment can enhance their homing and (iii) the molecular mechanisms accountable for any enhancement. Methods Adherent and free-flowing HSCs were determined in an intravital murine model of renal ischaemia-reperfusion injury. Some HSCs and animals were pre-treated prior to HSC infusion with function blocking antibodies, hyaluronidase or cytokines. Changes in surface expression and clustering of HSC adhesion molecules were determined using flow cytometry and confocal microscopy. HSC adhesion to endothelial counter-ligands (VCAM-1, hyaluronan) was determined using static adhesion assays in vitro. Results CD49d, CD44, VCAM-1 and hyaluronan governed HSC adhesion to the IR-injured kidney. Both KC and SDF-1α pre-treatment strategies significantly increased HSC adhesion within injured kidney, whilst SDF-1α also increased numbers continuing to circulate. SDF-1α and KC did not increase CD49d or CD44 expression but increased HSC adhesion to VCAM-1 and hyaluronan respectively. SDF-1α increased CD49d surface clustering, as well as HSC deformability. Conclusion Increasing HSC adhesive capacity for its endothelial counter-ligands, potentially through surface clustering, may explain their enhanced renal retention in vivo. Furthermore, increasing HSC deformability through SDF-1α treatment could explain the prolonged systemic circulation; the HSC can therefore continue to survey the damaged tissue instead of becoming entrapped within non-injured sites. Therefore manipulating these mechanisms of HSC recruitment outlined may improve the clinical outcome of cellular therapies for kidney disease.
Collapse
Affiliation(s)
- Rebecca L. White
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gerard Nash
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dean P. J. Kavanagh
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Caroline O. S. Savage
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neena Kalia
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Abstract
BACKGROUND Acute kidney injury (AKI) occurs in 26% of trauma patients and is associated with increased mortality and risk for nosocomial infections (NCIs). We compared serial plasma cytokine levels in patients with posttraumatic AKI to determine whether the early cytokine changes are associated with the occurrence of AKI and NCI. METHODS We performed a secondary analysis of the Inflammation and the Host Response to Injury database to include adult blunt trauma patients who had available plasma proteomic analyses. AKI was defined by the RIFLE (Risk, Injury, Failure, Loss, and End-stage Kidney) classification, which requires a 50% increase in serum creatinine. The association among AKI, NCI, and plasma cytokines was analyzed using a mixed model analyses and logistic regression. RESULTS Among 147 patients in the cohort, prevalence of NCI was 73% and 52% for patients with and without AKI, respectively. In mixed model analyses adjusted for clinical factors, AKI patients developed significant early increase in IL-1ra, IL-8, MCP1, and IL-6; early decrease in sTNFR2; and late decrease in IL-1ra, IL-4, and IL-6 concentrations, compared with patients without AKI and regardless of NCI. The change in cytokine pattern differed for sIL1R2, CXCL1, and MIP1β, depending on the occurrence of NCI. Patients with AKI and NCI had lower early and late sIL1R2 and higher early and late CXCL1 and MIP1β levels. Within the first 24 hours of injury, adding plasma levels of IL-1ra, IL-8, MCP1, IL-6, and sTNFR2 to clinical parameters of injury severity provided a predictive model for AKI superior to clinical model only (p < 0.001). CONCLUSION AKI trauma patients exhibit simultaneous changes in proinflammatory and anti-inflammatory serial plasma cytokine levels. The predictive model for AKI that combines plasma cytokine levels with clinical data within 24 hours of injury requires further prospective validation in larger studies. LEVEL OF EVIDENCE Prognostic study, level III.
Collapse
|
24
|
Abstract
BACKGROUND Acute kidney injury (AKI) is a common and serious problem in critically ill patients. Tests currently used to detect AKI (i.e., serum creatinine, serum urea and various urinary indices) often result in serious delays in detection of clinically relevant injury. This delayed detection translates into a potential missed opportunity for therapeutic interventions at a time when kidney damage may be limitable or reversible. This is also recognized as a potential reason for the poor clinical outcomes often associated with AKI. OBJECTIVES To appraise the recent literature characterizing several novel serum and urinary biomarkers, including neutrophil gelatinase-associated lipocalin, IL-18 and kidney injury molecule-1, which are capable of detecting AKI at an earlier phase of injury. Also to discuss the pitfalls of current conventional testing in kidney injury. METHOD Narrative literature review. CONCLUSIONS These novel biomarkers can detect injury when damage may still be reversible, allow for early risk stratification and/or prognostication, and are associated in early clinical studies with important outcomes such as severity of AKI, need for renal replacement therapy and survival. There is optimism that these novel biomarkers will discriminate the underlying pathophysiology of AKI (i.e., ischemia, sepsis, toxins or multifactorial), discriminate AKI from other renal disease (i.e., chronic kidney disease) and aid in localizing the site of acute injury in the kidney. As such, the future may entail development of an 'AKI biomarker panel' (i.e., analogous to a cardiac or liver enzyme panel) for use in clinical practice.
Collapse
Affiliation(s)
- Sean M Bagshaw
- University of Alberta Hospital, 3C1.12 Walter C. Mackenzie Centre, Division of Critical Care Medicine, 8440-112 Street, Edmonton, Alberta, T6G2B7, Canada +1 780 407 6755 ; +1 780 407 1228 ;
| |
Collapse
|
25
|
Abstract
Acute kidney injury (AKI) often results from ischemia reperfusion, sepsis, or exposure to nephrotoxins and is associated with a high rate of mortality and morbidity. Advances in understanding the pathophysiology of AKI may lead to the development of specific therapies. Although there is evidence of an important role for immune cells in AKI, the specific relevant populations and the mechanisms of their actions are unclear. In this issue of the JCI, Li et al. demonstrate that adenosine manipulates DC responses to kidney injury, raising hope that immunotherapy could be a tangible approach to AKI.
Collapse
Affiliation(s)
- Hamid Rabb
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
26
|
Xu X, Kriegel AJ, Liu Y, Usa K, Mladinov D, Liu H, Fang Y, Ding X, Liang M. Delayed ischemic preconditioning contributes to renal protection by upregulation of miR-21. Kidney Int 2012; 82:1167-75. [PMID: 22785173 PMCID: PMC3777822 DOI: 10.1038/ki.2012.241] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Delayed ischemic preconditioning effectively protects kidneys from ischemia-reperfusion injury but the mechanism underlying renal protection remains poorly understood. Here we examined the in vivo role of microRNA miR-21 in the renal protection conferred by delayed ischemic preconditioning in mice. A 15 minute renal ischemic preconditioning significantly increased the expression of miR-21 by 4 hours and substantially attenuated ischemia-reperfusion injury induced 4 days later. A locked nucleic acid-modified anti-miR-21 given at the time of ischemic preconditioning knocked down miR-21 and significantly exacerbated subsequent ischemia-reperfusion injury in the mouse kidney. Knockdown of miR-21 resulted in significant upregulation of programmed cell death protein 4, a pro-apoptotic target gene of miR-21, and substantially increased tubular cell apoptosis. Hypoxia inducible factor-1α in the kidney was activated after ischemic preconditioning and blockade of its activity with a decoy abolished the up-regulation of miR-21 in cultured human renal epithelial cells treated with the inducer cobalt chloride. In the absence of ischemic preconditioning, knockdown of miR-21 alone did not significantly affect ischemia-reperfusion injury in the mouse kidney. Thus, upregulation of miR-21 contributes to the protective effect of delayed ischemic preconditioning against subsequent renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xialian Xu
- Division of Nephrology, Shanghai Medical College, Fudan University, Zhongshan Hospital, Shanghai, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Fukudome EY, Li Y, Kochanek AR, Lu J, Smith EJ, Liu B, Kim K, Velmahos GC, deMoya MA, Alam HB. Pharmacologic resuscitation decreases circulating cytokine-induced neutrophil chemoattractant-1 levels and attenuates hemorrhage-induced acute lung injury. Surgery 2012; 152:254-61. [PMID: 22657731 DOI: 10.1016/j.surg.2012.03.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Accepted: 03/09/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Acute lung injury (ALI) is a complication of hemorrhagic shock (HS). Histone deacetylase inhibitors, such as valproic acid (VPA), can improve survival after HS; however, their effects on late organ injury are unknown. Herein, we have investigated the effects of HS and VPA treatment on ALI and circulating cytokines that may serve as biomarkers for the development of organ injury. METHODS Anesthetized Wistar-Kyoto rats (250-300 g) underwent 40% blood volume hemorrhage over 10 minutes followed by 30 minutes of unresuscitated shock and were treated with either VPA (300 mg/kg) or vehicle control. Blood samples were obtained at baseline, after shock, and before death (at 1, 4, and 20 hours; n = 3-4/timepoint/group). Serum samples were screened for possible biomarkers using a multiplex electrochemiluminescence detection assay, and results were confirmed using enzyme-linked immunosorbent assay (ELISA). In addition, lung tissue lysate was examined for chemokine and myeloperoxidase (MPO) levels as a marker for neutrophil infiltration and ALI. Lung cytokine-induced neutrophil chemoattractant-1 (CINC-1; a chemokine belonging to the interleukin-8 family that promotes neutrophil chemotaxis) mRNA levels were measured by real-time polymerase chain reaction studies. RESULTS Serum screening revealed that hemorrhage rapidly altered levels of circulating CINC-1. ELISA confirmed that CINC-1 protein was significantly elevated in the serum as early as 4 hours and in the lung at 20 hours after hemorrhage, without any significant changes in CINC-1 mRNA expression. Lung MPO levels were also elevated at both 4 and 20 hours after hemorrhage. VPA treatment attenuated these changes. CONCLUSION Hemorrhage resulted in the development of ALI, which was prevented with VPA treatment. Circulating CINC-1 levels rose rapidly after hemorrhage, and serum CINC-1 levels correlated with lung CINC-1 and MPO levels. This suggests that circulating CINC-1 levels could be used as an early marker for the subsequent development of organ inflammation and injury.
Collapse
Affiliation(s)
- Eugene Y Fukudome
- Department of Surgery, Division of Trauma, Emergency Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Renoprotective Effects of AVE0991, a Nonpeptide Mas Receptor Agonist, in Experimental Acute Renal Injury. Int J Hypertens 2012; 2012:808726. [PMID: 22319645 PMCID: PMC3272821 DOI: 10.1155/2012/808726] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 10/21/2011] [Indexed: 11/29/2022] Open
Abstract
Renal ischemia and reperfusion (I/R) is the major cause of acute kidney injury in hospitalized patients. Mechanisms underlying reperfusion-associated injury include recruitment and activation of leukocytes and release of inflammatory mediators. In this study, we investigated the renal effects of acute administration of AVE0991, an agonist of Mas, the angiotensin-(1–7) receptor, the angiotensin-(1–7) receptor, in a murine model of renal I/R. Male C57BL/6 wild-type or Mas−/− mice were subjected to 30 min of bilateral ischemia and 24 h of reperfusion. Administration of AVE0991 promoted renoprotective effects, as seen by improvement of function, decreased tissue injury, prevention of local and remote leucocyte infiltration, and release of the chemokine, CXCL1. I/R injury was similar in WT and Mas−/− mice, suggesting that endogenous activation of this receptor does not control renal damage under baseline conditions. In conclusion, pharmacological interventions using Mas receptor agonists may represent a therapeutic opportunity for the treatment of renal I/R injury.
Collapse
|
29
|
Rosner MH, Ronco C, Okusa MD. The Role of Inflammation in the Cardio-Renal Syndrome: A Focus on Cytokines and Inflammatory Mediators. Semin Nephrol 2012; 32:70-8. [DOI: 10.1016/j.semnephrol.2011.11.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Reverse signaling through the costimulatory ligand CD137L in epithelial cells is essential for natural killer cell-mediated acute tissue inflammation. Proc Natl Acad Sci U S A 2011; 109:E13-22. [PMID: 22160719 DOI: 10.1073/pnas.1112256109] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Renal ischemia-reperfusion injury (IRI) after kidney transplantation is a major cause of delayed graft function. Even though IRI is recognized as a highly coordinated and specific process, the pathways and mechanisms through which the innate response is activated are poorly understood. In this study, we used a mouse model of acute kidney IRI to examine whether the interactions of costimulatory receptor CD137 and its ligand (CD137L) are involved in the early phase of acute kidney inflammation caused by IRI. We report here that the specific expressions of CD137 on natural killer cells and of CD137L on tubular epithelial cells (TECs) are required for acute kidney IRI. Reverse signaling through CD137L in TECs results in their production of the chemokine (C-X-C motif) receptor 2 ligands CXCL1 and CXCL2 and the subsequent induction of neutrophil recruitment, resulting in a cascade of proinflammatory events during kidney IRI. Our findings identify an innate pathogenic pathway for renal IRI involving the natural killer cell-TEC-neutrophil axis, whereby CD137-CD137L interactions provide the causal contribution of epithelial cell dysregulation to renal IRI. The CD137L reverse signaling pathway in epithelial cells therefore may represent a good target for blocking the initial stage of inflammatory diseases, including renal IRI.
Collapse
|
31
|
Faleiros RR, Leise BS, Watts M, Johnson PJ, Black SJ, Belknap JK. Laminar chemokine mRNA concentrations in horses with carbohydrate overload-induced laminitis. Vet Immunol Immunopathol 2011; 144:45-51. [DOI: 10.1016/j.vetimm.2011.07.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/13/2011] [Accepted: 07/08/2011] [Indexed: 01/14/2023]
|
32
|
Wan X, Yang J, Xing L, Fan L, Hu B, Chen X, Cao C. Inhibition of IκB Kinase β attenuates hypoxia-induced inflammatory mediators in rat renal tubular cells. Transplant Proc 2011; 43:1503-10. [PMID: 21693225 DOI: 10.1016/j.transproceed.2011.01.179] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 01/11/2011] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Inflammation is now believed to play a major role in the pathophysiology of ischemic acute kidney injury (AKI), which is thought to be directly regulated by nuclear factor-κB (NF-κB). Our previous study indicated that ischemic preconditioning (IPC) alleviated renal ischemic-reperfusion injury due to inhibition of IκB kinase β (IKK β) activity. Using small interfering RNA (siRNA) to silence the expression of IKKβ, which consists of the IKK complex residing at a key convergence site that leads to NF-κB activation in multiple signaling pathways, we protected organs from ischemic AKI. Herein, we have report a siRNA-based treatment to prevent ischemic AKI. METHODS Ischemic AKI was induced by a hypoxia-mimicking agent cobalt chloride (CoCl(2)). The therapeutic effects of IKKβ-specific siRNA were evaluated on the expression of interleukin (IL)-18, neutrophil gelatinase-associated lipocalin (NGAL), and cell apoptosis. RESULTS Compared with CoCl(2)-induced NRK52E cells, pretransfected IKKβ-specific siRNA reduced the expression of IL-18 and NGAL to 62.5% and 50.4% in messenger RNA (mRNA) and to 57.2% and 62.7% in protein levels, respectively. The necrosis index in the IKKβ-specific siRNA transfected group was decreased compared with a nonspecific siRNA transfected group. CONCLUSIONS These data revealed that hypoxia-induced inflammatory responses were IKKβ/NF-κB-dependent. Knockdown of IKKβ by siRNA suppressed the transcription IKKβ/NF-κB-mediated inflammatory mediators in tumor necrosis factor-α or CoCl(2)-treated tubular epithelial cells, and decreased CoCl(2)-induced cell death, which may be a useful, preventive and therapeutic strategy for ischemic AKI.
Collapse
Affiliation(s)
- X Wan
- Department of Nephrology, Nanjing First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Rajan D, Wu R, Shah KG, Jacob A, Coppa GF, Wang P. Human ghrelin protects animals from renal ischemia-reperfusion injury through the vagus nerve. Surgery 2011; 151:37-47. [PMID: 21943641 DOI: 10.1016/j.surg.2011.06.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 06/16/2011] [Indexed: 02/03/2023]
Abstract
BACKGROUND Acute kidney injury secondary to renal ischemia and reperfusion injury is widely prevalent. Ghrelin, which is a stomach-derived peptide, has been shown to be anti-inflammatory. The purpose of this study was to examine whether human ghrelin has any beneficial effects after renal ischemia and reperfusion injury, and if so, whether ghrelin's action in renal ischemia and reperfusion injury is mediated by the vagus nerve. METHODS Male adult rats were subjected to renal ischemia and reperfusion by bilateral renal pedicle clamping for 60 min, treated intravenously with human ghrelin (4 nmol/rat) or normal saline (vehicle) immediately after reperfusion. After 24 h, the animals were killed and samples were harvested. In separate groups, subdiaphragmatic vagotomy prior to renal ischemia and reperfusion was performed, treated with human ghrelin or vehicle, and at 24 h, blood and organs were harvested. RESULTS Renal ischemia and reperfusion injury caused significant increases in the serum levels of tissue injury markers compared with the sham operation. Human ghrelin treatment attenuated serum creatinine and blood urea nitrogen significantly by 55% and 53%, and liver enzymes (aminotransferase [AST] and alanine aminotransferase [ALT]) by 20% and 24%, respectively, compared with the vehicle-treated groups. Tissue water contents, plasma and kidney interleukin-6, and kidney myeloperoxidase activity were decreased. Bcl-2/Bax ratio was increased, and histology of the kidneys was improved. More importantly, prior vagotomy abolished ghrelin's protective effect in tissue injury markers and tissue water contents in renal ischemia and reperfusion injured animals. CONCLUSION Human ghrelin treatment in renal ischemia and reperfusion injured rats attenuated systemic and kidney-specific inflammatory responses. The protection of human ghrelin in renal ischemia and reperfusion injury was mediated by the vagus nerve. These data suggest that ghrelin can be developed as a novel treatment for patients with acute kidney injury induced by renal ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Derry Rajan
- Department of Surgery, North Shore University Hospital and Long Island Jewish Medical Center, Manhasset, NY 11030, USA
| | | | | | | | | | | |
Collapse
|
34
|
Role of Nrf2 in host defense against influenza virus in cigarette smoke-exposed mice. J Virol 2011; 85:4679-90. [PMID: 21367886 DOI: 10.1128/jvi.02456-10] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza virus is a common respiratory tract viral infection. Although influenza can be fatal in patients with chronic pulmonary diseases such as chronic obstructive pulmonary disease, its pathogenesis is not fully understood. The Nrf2-mediated antioxidant system is essential to protect the lungs from oxidative injury and inflammation. In the present study, we investigated the role of Nrf2 in protection against influenza virus-induced pulmonary inflammation after cigarette smoke exposure with both in vitro and in vivo approaches. For in vitro analyses, peritoneal macrophages isolated from wild-type and Nrf2-deficient mice were treated with poly(I:C) and/or cigarette smoke extract. For in vivo analysis, these mice were infected with influenza A virus with or without exposure to cigarette smoke. In Nrf2-deficient macrophages, NF-κB activation and the induction of its target inflammatory genes were enhanced after costimulation with cigarette smoke extract and poly(I:C) compared with wild-type macrophages. The induction of antioxidant genes was observed for the lungs of wild-type mice but not those of Nrf2-deficient mice after cigarette smoke exposure. Cigarette smoke-exposed Nrf2-deficient mice showed higher rates of mortality than did wild-type mice after influenza virus infection, with enhanced peribronchial inflammation, lung permeability damage, and mucus hypersecretion. Lung oxidant levels and NF-κB-mediated inflammatory gene expression in the lungs were also enhanced in Nrf2-deficient mice. Our data indicate that the antioxidant pathway controlled by Nrf2 is pivotal for protection against the development of influenza virus-induced pulmonary inflammation and injury under oxidative conditions.
Collapse
|
35
|
Stokman G, Stroo I, Claessen N, Teske GJD, Weening JJ, Leemans JC, Florquin S. Stem cell factor expression after renal ischemia promotes tubular epithelial survival. PLoS One 2010; 5:e14386. [PMID: 21200435 PMCID: PMC3006174 DOI: 10.1371/journal.pone.0014386] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 11/30/2010] [Indexed: 11/18/2022] Open
Abstract
Background Renal ischemia leads to apoptosis of tubular epithelial cells and results in decreased renal function. Tissue repair involves re-epithelialization of the tubular basement membrane. Survival of the tubular epithelium following ischemia is therefore important in the successful regeneration of renal tissue. The cytokine stem cell factor (SCF) has been shown to protect the tubular epithelium against apoptosis. Methodology/Principal Findings In a mouse model for renal ischemia/reperfusion injury, we studied how expression of c-KIT on tubular epithelium and its ligand SCF protect cells against apoptosis. Administration of SCF specific antisense oligonucleotides significantly decreased specific staining of SCF following ischemia. Reduced SCF expression resulted in impaired renal function, increased tubular damage and increased tubular epithelial apoptosis, independent of inflammation. In an in vitro hypoxia model, stimulation of tubular epithelial cells with SCF activated survival signaling and decreased apoptosis. Conclusions/Significance Our data indicate an important role for c-KIT and SCF in mediating tubular epithelial cell survival via an autocrine pathway.
Collapse
Affiliation(s)
- Geurt Stokman
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
36
|
Dennen P, Altmann C, Kaufman J, Klein CL, Andres-Hernando A, Ahuja NH, Edelstein CL, Cadnapaphornchai MA, Keniston A, Faubel S. Urine interleukin-6 is an early biomarker of acute kidney injury in children undergoing cardiac surgery. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:R181. [PMID: 20942931 PMCID: PMC3219287 DOI: 10.1186/cc9289] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 09/02/2010] [Accepted: 10/13/2010] [Indexed: 11/21/2022]
Abstract
Introduction Interleukin-6 (IL-6) is a proinflammatory cytokine that increases early in the serum of patients with acute kidney injury (AKI). The aim of this study was to determine whether urine IL-6 is an early biomarker of AKI and determine the source of urine IL-6. Numerous proteins, including cytokines, are filtered by the glomerulus and then endocytosed and metabolized by the proximal tubule. Since proximal tubule injury is a hallmark of AKI, we hypothesized that urine IL-6 would increase in AKI due to impaired proximal tubule metabolism of filtered IL-6. Methods Urine was collected in 25 consecutive pediatric patients undergoing cardiac bypass surgery (CPB). AKI was defined as a 50% increase in serum creatinine at 24 hours (RIFLE (Risk, Injury, Failure, Loss, End stage), R). Mouse models of AKI and freshly isolated proximal tubules were also studied. Results Urine IL-6 increased at six hours in patients with AKI versus no AKI (X2 = 8.1750; P < 0.0042). Urine IL-6 > 75 pg/mg identified AKI with a sensitivity of 88%. To assess whether increased urine IL-6 occurs in functional versus structural renal failure, mouse models of pre-renal azotemia after furosemide injection (no tubular injury), ischemic AKI (tubular injury) and cisplatin AKI (tubular injury) were studied. Urine IL-6 did not significantly increase in pre-renal azotemia but did increase in ischemic and cisplatin AKI. To determine if circulating IL-6 appears in the urine in AKI, recombinant human (h)IL-6 was injected intravenously and urine collected for one hour; urine hIL-6 increased in ischemic AKI, but not pre-renal azotemia. To determine the effect of AKI on circulating IL-6, serum hIL-6 was determined one hour post-intravenous injection and was increased in ischemic AKI, but not pre-renal azotemia. To directly examine IL-6 metabolism, hIL-6 was added to the media of normal and hypoxic isolated proximal tubules; hIL-6 was reduced in the media of normal versus injured hypoxic proximal tubules. Conclusions Urine IL-6 increases early in patients with AKI. Animal studies demonstrate that failure of proximal tubule metabolism of IL-6 results in increased serum and urine IL-6. Impaired IL-6 metabolism leading to increased serum IL-6 may contribute to the deleterious systemic effects and increased mortality associated with AKI.
Collapse
Affiliation(s)
- Paula Dennen
- Department of Medicine, Division of Nephrology, Denver Health & Hospitals, 777 Bannock Street, Denver, CO 80204, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Tadagavadi RK, Reeves WB. Endogenous IL-10 attenuates cisplatin nephrotoxicity: role of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:4904-11. [PMID: 20844196 DOI: 10.4049/jimmunol.1000383] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sterile inflammation is associated with tissue injury and organ failure. Recent studies indicate that certain endogenous cytokines and immune cells may limit tissue injury by reducing immune-mediated inflammatory responses. Cisplatin is a commonly used anticancer chemotherapeutic agent but causes acute kidney injury and dysfunction. In a recent study, we showed that renal dendritic cells attenuate cisplatin-induced kidney injury by reducing inflammation. In this study, we investigated the effect of endogenous IL-10 and dendritic cell IL-10 in cisplatin-mediated kidney injury. Cisplatin treatment caused increases in renal IL-10R1 expression and STAT3 phosphorylation. In response to cisplatin treatment, IL-10 knockout mice showed more rapid and greater increases in blood urea nitrogen and serum creatinine compared with wild-type mice, indicating that endogenous IL-10 ameliorates kidney injury in cisplatin nephrotoxicity. Renal infiltration of IFN-γ-producing neutrophils was markedly increased in IL-10 knockout mice compared with wild-type mice. However, IFN-γ neutralization had no impact on renal dysfunction, suggesting IFN-γ-independent mechanisms of tissue injury in cisplatin nephrotoxicity. Renal dendritic cells showed high expression of IL-10 in response to cisplatin treatment. We further investigated the effect of dendritic cell-derived IL-10 in cisplatin nephrotoxicity using a conditional cell ablation approach. Mixed bone marrow chimeric mice lacking IL-10 in dendritic cells showed moderately greater renal dysfunction than chimeric mice positive for IL-10 in dendritic cells. These data demonstrate that endogenous IL-10 reduces cisplatin nephrotoxicity and associated inflammation. Moreover, IL-10 produced by dendritic cells themselves accounts for a portion of the protective effect of dendritic cells in cisplatin nephrotoxicity.
Collapse
|
38
|
McDuffie JE, Sablad M, Ma J, Snook S. Urinary parameters predictive of cisplatin-induced acute renal injury in dogs. Cytokine 2010; 52:156-62. [PMID: 20655244 DOI: 10.1016/j.cyto.2010.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 02/15/2010] [Accepted: 06/02/2010] [Indexed: 11/16/2022]
Abstract
A 28-day study was conducted to evaluate changes in urinary cytokine/chemokine expression levels in dogs with renal injury due to administration of cisplatin. Animals (n=17) were administered cisplatin at 0.75 mg/kg/day (i.v.) for five consecutive days. Urine/serum were collected at pre-dosing, 4h post-dosing and on days 2, 3, 4, 8, 10, 14, 16, 18, 21, 23, 25, 28 and unscheduled terminations. Animals were euthanized when serum creatinine (sCr) levels measured at ≥ 1.9 mg/dL, indicating significant loss of renal function (decreased glomerular filtration rate). Relevant clinical observations included lethargy and dehydration. Pre-study sCr levels ranged from 0.6 to 0.8 mg/dL; on days 1 through 4, sCr levels ranged from 0.5 and 1.1mg/dL; and terminal sCr levels ranged from 0.6 and 6.6 mg/dL. Histologically, cisplatin-related renal changes were characterized as proximal tubule dilatation, vacuolization, degeneration, regeneration, and interstitial inflammation. Increased interleukin (IL)-2, IL-8, monocyte chemoattractant protein-1 (MCP-1), granulocyte-macrophage colony-stimulating factor (GMCSF) and keratinocyte-derived chemokine (KC) occurred on days 3 through 4. Increased IL-7 occurred on day 4. This study showed for the first time that inflammatory cytokines/chemokines in urine positively identified acute renal tubular injury in dogs at time points earlier than sCr, a traditional marker of nephrotoxicity.
Collapse
Affiliation(s)
- J Eric McDuffie
- Global Preclinical Development, Johnson & Johnson Pharmaceutical Research and Development LLC, 3210 Merryfield Row, San Diego, CA 92121, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Biological modulation of renal ischemia-reperfusion injury holds the potential to reduce the incidence of early graft dysfunction and to safely expand the donor pool with kidneys that have suffered prolonged ischemic injury before organ recovery. RECENT FINDINGS In the current review, we will discuss clinical studies that compare kidney transplant recipients with and without early graft dysfunction in order to elucidate the pathophysiology of ischemic acute allograft injury. We will specifically review the mechanisms leading to depression of the glomerular filtration rate and activation of the innate immune system in response to tissue injury. SUMMARY We conclude that the pathophysiology of delayed graft function after kidney transplantation is complex and shares broad similarity with rodent models of ischemic acute kidney injury. Given the lack of specific therapies to prevent delayed graft function in transplant recipients, comprehensive efforts should be initiated to translate the promising findings obtained in small animal models into clinical interventions that attenuate ischemic acute kidney injury after transplantation.
Collapse
|
40
|
Kim M, Park SW, Kim M, Chen SWC, Gerthoffer WT, D'Agati VD, Lee HT. Selective renal overexpression of human heat shock protein 27 reduces renal ischemia-reperfusion injury in mice. Am J Physiol Renal Physiol 2010; 299:F347-58. [PMID: 20484296 DOI: 10.1152/ajprenal.00194.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
We have previously shown that exogenous and endogenous A(1) adenosine receptor (A(1)AR) activation protected against renal ischemia-reperfusion (IR) injury in mice by induction and phosphorylation of heat shock protein 27 (HSP27). With global overexpression of HSP27 in mice, however, there was a paradoxical increase in systemic inflammation with increased renal injury after an ischemic insult due to increased NK1.1 cytotoxicity. In this study, we hypothesized that selective renal expression of HSP27 in mice would improve renal function and reduce injury after IR. Mice were subjected to renal IR injury 2 days after intrarenal injection of saline or a lentiviral construct encoding enhanced green fluorescent protein (EGFP) or human HSP27 coexpressing EGFP (EGFP-huHSP27). Mice with kidney-specific reconstitution of huHSP27 had significantly lower plasma creatinine, renal necrosis, apoptosis, and inflammation as demonstrated by decreased proinflammatory cytokine mRNA induction and neutrophil infiltration. In addition, there was better preservation of the proximal tubule epithelial filamentous (F)-actin cytoskeleton in the huHSP27-reconstituted groups than in the control groups. Furthermore, huHSP27 overexpression led to increased colocalization with F-actin in renal proximal tubules. Taken together, these findings have important clinical implications, as they imply that kidney-specific expression of HSP27 through lentiviral delivery is a viable therapeutic option in attenuating the effects of renal IR.
Collapse
Affiliation(s)
- Minjae Kim
- Departments of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York 10032-3784, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Soloski MJ, Crow MT, Rabb H. Mycophenolate mofetil modifies kidney tubular injury and Foxp3+ regulatory T cell trafficking during recovery from experimental ischemia-reperfusion. Transpl Immunol 2010; 23:45-52. [PMID: 20412855 DOI: 10.1016/j.trim.2010.04.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 03/02/2010] [Accepted: 04/05/2010] [Indexed: 01/25/2023]
Abstract
Lymphocytes participate in the early pathogenesis of ischemia-reperfusion injury (IRI) in kidney; however, their role during repair is largely unknown. Recent data have shown that Foxp3(+) regulatory T cells (Tregs) traffic into kidney during healing from IRI and directly participate in repair. Since lymphocyte-targeting therapy is currently administered to prevent rejection during recovery from IRI in renal transplants, we hypothesized that mycophenolate mofetil (MMF) would alter Treg trafficking and kidney repair. C57BL/6J and T cell deficient mice underwent unilateral clamping of renal pedicle for 45 min, followed by reperfusion, and were sacrificed at day 10. Mice were treated with saline (C) or MMF (100mg/kg) i.p. daily starting at day 2 until sacrifice (n=5-12/group). MMF worsened kidney tubular damage compared to C at 10 days (cortex and outer medulla: p<0.05) in wild-type mice; tubular apoptotic index was increased in cortex in MMF group as well (p=0.01). MMF reduced the total number of kidney-infiltrating mononuclear cells (p<0.001 versus C) and the percentages of TCRbeta(+)CD4(+) and TCRbeta(+)CD8(+) T cells (p<0.01), but not natural killer (NK), NKT or B lymphocytes. MMF specifically reduced kidney Foxp3(+) Tregs (0.82+/-0.11% versus 1.75+/-0.17%, p<0.05). Tubular proliferative index and tissue levels of basic FGF were increased in MMF group (p<0.05), IL-10 and IL-6 were decreased (p<0.05). To evaluate if MMF effect occurred through non-lymphocytic cells, T cell deficient mice were treated with MMF. Tubular injury in T cell deficient mice was not affected by MMF treatment, though MMF-treated animals had increased VEGF and decreased PDGF-BB protein tissue levels compared to controls (p<0.05). Thus, MMF modifies the structural, epithelial proliferative and inflammatory response during healing, likely through effects on T cells and possibly Tregs. Kidney repair after IRI can be altered by agents that target lymphocytes.
Collapse
Affiliation(s)
- Maria Teresa Gandolfo
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Preconditioning by toll-like receptor 2 agonist Pam3CSK4 reduces CXCL1-dependent leukocyte recruitment in murine myocardial ischemia/reperfusion injury. Crit Care Med 2010; 38:903-9. [PMID: 20081527 DOI: 10.1097/ccm.0b013e3181ce50e6] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To test whether preconditioning with a toll-like receptor (TLR) 2 agonist protects against myocardial ischemia and reperfusion by interfering with chemokine CXCL1 release from cardiomyocytes. DESIGN C3H mice were challenged with vehicle or synthetic TLR2 agonist Pam3Cys-Ser-Lys4 (Pam3CSK4; 1 mg/kg) 24 hrs before myocardial ischemia (20 mins) and reperfusion (2 hrs or 24 hrs). Infarct size, troponin T release, and leukocyte recruitment were quantified. In murine cardiomyocytes (HL-1), we studied the expression/activation profile of TLR2 in response to stimulation with Pam3CSK4 (0.01-1 mg/mL). Furthermore, we studied the chemokine ligand 1 (CXCL1) response to Pam3CSK4 and ischemia/reperfusion in vivo and in vitro. SETTING University hospital research laboratory. SUBJECTS Anesthetized male mice and murine cardiomyocytes. MEASUREMENTS AND MAIN RESULTS Preconditioning by Pam3CSK4 reduced infarct size and troponin T release. This was accompanied by a decreased recruitment of leukocytes into the ischemic area and an improved cardiac function. In HL-1 cells, TLR2 activation amplified the expression of the receptor in a time-dependent manner and led to CXCL1 release in a concentration-dependent manner. Preconditioning by Pam3CSK4 impaired CXCL1 release in response to a second inflammatory stimulus in vivo and in vitro. CONCLUSIONS Preconditioning by TLR2 agonist Pam3CSK4 reduces myocardial infarct size after myocardial ischemia/reperfusion. One of the mechanisms involved is a diminished chemokine release from cardiomyocytes, which subsequently limits leukocyte infiltration.
Collapse
|
43
|
Shigeoka AA, Kambo A, Mathison JC, King AJ, Hall WF, da Silva Correia J, Ulevitch RJ, McKay DB. Nod1 and nod2 are expressed in human and murine renal tubular epithelial cells and participate in renal ischemia reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2010; 184:2297-304. [PMID: 20124104 DOI: 10.4049/jimmunol.0903065] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nucleotide-binding oligomerization domain (Nod) 1 and Nod2 are members of a family of intracellular innate sensors that participate in innate immune responses to pathogens and molecules released during the course of tissue injury, including injury induced by ischemia. Ischemic injury to the kidney is characterized by renal tubular epithelial apoptosis and inflammation. Among the best studied intracellular innate immune receptors known to contribute to apoptosis and inflammation are Nod1 and Nod2. Our study compared and contrasted the effects of renal ischemia in wild-type mice and mice deficient in Nod1, Nod2, Nod(1 x 2), and in their downstream signaling molecule receptor-interacting protein 2. We found that Nod1 and Nod2 were present in renal tubular epithelial cells in both mouse and human kidneys and that the absence of these receptors in mice resulted in protection from kidney ischemia reperfusion injury. Significant protection from kidney injury was seen with a deficiency of Nod2 and receptor-interacting protein 2, and the simultaneous deficiency of Nod1 and Nod2 provided even greater protection. We conclude that the intracellular sensors Nod1 and Nod2 play an important role in the pathogenesis of acute ischemic injury of the kidney, although possibly through different mechanisms.
Collapse
Affiliation(s)
- Alana A Shigeoka
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Ischemia reperfusion injury (IRI) is a choreographed process leading to delayed graft function (DGF) and reduced long-term patency of the transplanted organ. Early identification of recipients of grafts at risk would allow modification of the posttransplant management, and thereby potentially improve short- and long-term outcomes. The recently emerged "omics" technologies together with bioinformatics workup have allowed the integration and analysis of IRI-associated molecular profiles in the context of DGF. Such a systems biological approach promises qualitative information about interdependencies of complex processes such as IRI regulation, rather than offering descriptive tables of differentially regulated features on a transcriptome, proteome, or metabolome level leaking the functional, biological framework. In deceased-donor kidney transplantation as the primary causative factor resulting in IRI and DGF, a distinct signature and choreography of molecular events in the graft before harvesting seems to be associated with subsequent DGF. A systems biological assessment of these molecular changes suggests that processes along inflammation are of pivotal importance for the early stage of IRI. The causal proof of this association has been tested by a double-blinded, randomized, controlled trial of steroid or placebo infusion into deceased donors before the organs were harvested. Thorough systems biological analysis revealed a panel of biomarkers with excellent discrimination. In summary, integrated analysis of omics data has brought forward biomarker candidates and candidate panels that promise early assessment of IRI. However, the clinical utility of these markers still needs to be established in prospective trials in independent patient populations.
Collapse
|
45
|
Liu M, Agreda P, Crow M, Racusen L, Rabb H. Effects of Delayed Rapamycin Treatment on Renal Fibrosis and Inflammation in Experimental Ischemia Reperfusion Injury. Transplant Proc 2009; 41:4065-71. [DOI: 10.1016/j.transproceed.2009.08.083] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 06/27/2009] [Accepted: 08/17/2009] [Indexed: 11/30/2022]
|
46
|
Sadeghi M, Daniel V, Naujokat C, Schmidt J, Mehrabi A, Zeier M, Opelz G. Decreasing plasma soluble IL-1 receptor antagonist and increasing monocyte activation early post-transplant may be involved in pathogenesis of delayed graft function in renal transplant recipients. Clin Transplant 2009; 24:415-23. [DOI: 10.1111/j.1399-0012.2009.01130.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Lee BH, Lee TJ, Jung JW, Oh DJ, Choi JC, Shin JW, Park IW, Choi BW, Kim JY. The role of keratinocyte-derived chemokine in hemorrhage-induced acute lung injury in mice. J Korean Med Sci 2009; 24:775-81. [PMID: 19794970 PMCID: PMC2752755 DOI: 10.3346/jkms.2009.24.5.775] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Accepted: 02/20/2009] [Indexed: 01/11/2023] Open
Abstract
Dominant inflammatory cytokines might be different depending on the underlying causes of acute lung injury (ALI). The role of kertinocyte-derived chemokine (KC), a potent chemoattractant for neutrophils, has not been clearly established in hemorrhage-induced ALI. In this study, lung injury and cytokine expression were evaluated in LPS- or hemorrhage-induced ALI models of BALB/c mice. The myeloperoxidase activities at 4 hr after hemorrhage and LPS-injection were 47.4+/-13.0 and 56.5+/-16.4 U/g, respectively. NF-kappaB activity peaked at 4 hr after hemorrhage, which was suppressed to the control level by anti-high mobility group B1 (HMGB1) antibody. Lung expressions of TNF-alpha, MIP-2, and IL-1beta were increased by LPS injection. However, there was only a minimal increase in IL-1beta and no expressions of TNF-alpha or MIP-2 in hemorrhage-induced ALI. In contrast, lung KC increased significantly at 4 hr after hemorrhage compared to control levels (83.1+/-12.3 vs. 14.2+/-1.6 pg/mL/mg by ELISA) (P<0.05). By immunohistochemical staining, lung neutrophils stained positive for KC. Increased KC was also observed in bronchoalveolar lavage fluid and plasma. KC plays an important role in hemorrhage-induced ALI.
Collapse
Affiliation(s)
- Byoung Hoon Lee
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Tae Jin Lee
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jae Woo Jung
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Dong Jin Oh
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jae Chol Choi
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jong Wook Shin
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - In Won Park
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Byoung Whui Choi
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jae Yeol Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Prasad S, McDaid JP, Tam FWK, Haylor JL, Ong ACM. Pkd2 dosage influences cellular repair responses following ischemia-reperfusion injury. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1493-503. [PMID: 19729489 DOI: 10.2353/ajpath.2009.090227] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) results from mutations in either PKD1 or PKD2 and accounts for 10% of all patients on renal replacement therapy. The kidney disease phenotype is primarily characterized by cyst formation, but there are also prominent interstitial changes (inflammation, apoptosis, proliferation, and fibrosis). Using a model of unilateral ischemia-reperfusion injury, we tested the hypothesis that Pkd2 heterozygous kidneys are more sensitive to injury and that this could lead to interstitial inflammation and fibrosis. Baseline tubular proliferation in heterozygous kidneys was twofold higher than in wild-type kidneys. The magnitude and duration of tubular and interstitial proliferative responses was consistently greater in injured heterozygous compared with wild-type kidneys at all time points. Conversely, tubular p21 expression in heterozygotes was lower at baseline and following injury at all time points. Significantly more neutrophils and macrophages were detected in injured Pkd2 heterozygous kidneys at 2 days, correlating with increased expression of the cytokines interleukin (IL)-1beta and keratinocyte-derived chemokine and resulting in interstitial fibrosis at 28 days. We conclude that Pkd2 dosage influences both susceptibility and nature of the repair responses following injury. Polycystin-2 is therefore likely to play multiple roles in regulating tubular cell viability, repair, and remodeling in the mature kidney.
Collapse
Affiliation(s)
- Sony Prasad
- Kidney Genetics Group, Academic Unit of Nephrology, Sheffield Kidney Institute, The Henry Wellcome Laboratories for Medical Research, University of Sheffield Medical School, Sheffield S10 2RX, UK
| | | | | | | | | |
Collapse
|
49
|
Faleiros R, Leise B, Westerman T, Yin C, Nuovo G, Belknap J. In Vivo and In Vitro Evidence of the Involvement of CXCL1, a Keratinocyte-Derived Chemokine, in Equine Laminitis. J Vet Intern Med 2009; 23:1086-96. [DOI: 10.1111/j.1939-1676.2009.0349.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
50
|
Gong W, Klöpfel M, Reutzel-Selke A, Jurisch A, Vogt K, Haase S, Höflich C, Polenz D, Gerstmayer B, Tomiuk S, Volk HD, Pascher A, Sawitzki B. High weight differences between donor and recipient affect early kidney graft function--a role for enhanced IL-6 signaling. Am J Transplant 2009; 9:1742-51. [PMID: 19563340 DOI: 10.1111/j.1600-6143.2009.02725.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The frequency of delayed function of kidney transplants varies greatly and is associated with quality of graft, donor age and the duration of cold ischemia time. Furthermore, body weight differences between donor and recipient can affect primary graft function, but the underlying mechanism is poorly understood. We transplanted kidney grafts from commensurate body weight (L-WD) or reduced body weight (H-WD) donor rats into syngeneic or allogeneic recipients. Twenty-four hours posttransplantation, serum creatinine levels in H-WD recipients were significantly higher compared to L-WD recipients indicating impaired primary graft function. This was accompanied by upregulation of IL-6 transcription and increased tubular destruction in grafts from H-WD recipients. Using DNA microarray analysis, we detected decreased expression of genes associated with kidney function and an upregulation of other genes such as Cyp3a13, FosL and Trib3. A single application of IL-6 into L-WD recipients is sufficient to impair primary graft function and cause tubular damage, whereas immediate neutralization of IL-6 receptor signaling in H-WD recipients rescued primary graft function with well-preserved kidney graft architecture and a normalized gene expression profile. These findings have strong clinical implication as anti-IL6R treatment of patients receiving grafts from lower-weight donors could be used to improve primary graft function.
Collapse
Affiliation(s)
- W Gong
- Institute of Medical Immunology, Charité University Medicine, Campus Mitte, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|