1
|
5-Methyltetrahydrofolate Attenuates Oxidative Stress and Improves Kidney Function in Acute Kidney Injury through Activation of Nrf2 and Antioxidant Defense. Antioxidants (Basel) 2022; 11:antiox11061046. [PMID: 35739943 PMCID: PMC9219715 DOI: 10.3390/antiox11061046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress is a major mediator of adverse outcomes in acute kidney injury (AKI). Deficiency of micronutrients, such as folate, is common in AKI. Our previous study reported that AKI impaired kidney reabsorption of folate and decreased plasma folate level in rats. The present study investigated the effect of 5-methyltetrahydrofolate (5-MTHF), a biologically active form of folate/folic acid, on AKI-impaired kidney function and oxidative stress. Sprague-Dawley rats developed AKI after kidney ischemia (45 min) and reperfusion (24 h). Injection of 5-MTHF (3 µg/kg body weight) improved kidney function and attenuated oxidative stress with a restoration of glutathione and a reduction of lipid peroxidation in the kidney. Injection of 5-MTHF activated transcription factor Nrf2 and increased the expression of glutathione synthesizing enzymes, superoxide dismutase-1 and heme oxygenase-1 in the kidney. Simulated ischemia-reperfusion through hypoxia-reoxygenation increased oxidative stress in proximal tubular cells. Incubation of cells with 5-MTHF alleviated cell injury and increased antioxidant enzyme expression and intracellular glutathione levels. Inhibition of Nrf2 expression through siRNA transfection abolished the effect of 5-MTHF against oxidative stress. These results suggest that low-dose folic acid can improve kidney function through activation of Nrf2 and restoration of antioxidant defence. Micronutrient supplements may improve clinical outcomes in AKI.
Collapse
|
2
|
Xue J, Zhu K, Cao P, Long C, Deng Y, Liu T, Yin G, Li X, Wang Z. Ischemic preconditioning-induced protective effect for promoting angiogenesis in renal ischemia-reperfusion injury by regulating miR-376c-3p/HIF-1α/VEGF axis in male rats. Life Sci 2022; 299:120357. [PMID: 35092734 DOI: 10.1016/j.lfs.2022.120357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 01/23/2022] [Accepted: 01/23/2022] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Ischemic preconditioning (IPC) is defined as a well-established phenomenon in which brief exposure to sublethal episodes of ischemia and reperfusion induces a tolerance to injurious effects of prolonged ischemia by exploiting intrinsic defence mechanisms. The present study was performed to determine the protective effect of IPC on the rat renal ischemia-reperfusion injury (IRI) via miR-376c-3p/HIF-1α/VEGF axis. METHODS In vivo, these male Sprague-Dawley rats were treated by IRI and IPC. Meanwhile, these rats from different treatment groups were also injected subcutaneously with 2 nmol agomir-376c-3p and/or 10 nmol recombinant rat HIF-1α. At 72 h after reperfusion, serum samples were respectively collected for renal function. Besides, kidney tissues were harvested to observe renal morphology changes. Subsequently, the expression levels of CD31, HIF-1α and VEGF in the kidney tissues were measured using immunohistochemical staining, quantitative real-time PCR, as well as Western blotting analysis at the indicated time points after reperfusion. In vitro, HK-2 cells were used to detect the cell activity by CCK-8 and transfection of mir-376c-3p mimic in Hypoxia/Reoxygenation (H/R) group. RESULTS In vivo, the pathological changes were significantly relieved in the rats with IPC group, compared to the IRI group. Rats which were treated IPC significantly reduced the levels of blood urea nitrogen (BUN), serum creatinine (Scr) at 24 h after operation, compared to IRI group. After IPC treatment, the expression level of CD31 was obviously decreased, compared to IRI group. A total of differently expressed microRNAs were screened out by microRNA microarray assay in rat renal ischemia tissue, especially showing that miR-376c-3p was selected as the target miRNA. Compared to IRI group, the expression level of miR-376c-3p were obviously higher in IPC-treated group. Double-luciferase reporter assay demonstrated that miR-376c-3p directly targeted HIF-1α. In vitro, IPC significantly increased cell viability of HK-2, and promoted the angiogenesis via up-regulating miR-376c-3p/HIF-1α/VEGF axis. Furthermore, the expression level of HIF-1α was apparently decreased in HK-2 treated with H/R after miR-376c-3p mimic transfection respectively, as well as the increased expression level of VEGF. CONCLUSIONS Our study provided a novel insight for investigating the protective effect of IPC on renal IRI. Consequently, miR-376c-3p played an important role in renal IRI by promoting angiogenesis via targeting HIF-1α/VEGF pathway in male rats.
Collapse
Affiliation(s)
- Jianxin Xue
- Department of Urology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Kai Zhu
- Department of Urology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Pu Cao
- Department of Urology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Chengcheng Long
- Department of Urology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Youming Deng
- Department of Anesthesiology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Tieshi Liu
- Department of Urology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Guoping Yin
- Department of Anesthesiology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Xiao Li
- Department of Urology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China.
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
3
|
Ribeiro A, Dobosz E, Krill M, Köhler P, Wadowska M, Steiger S, Schmaderer C, Koziel J, Lech M. Macrophage-Specific MCPIP1/Regnase-1 Attenuates Kidney Ischemia-Reperfusion Injury by Shaping the Local Inflammatory Response and Tissue Regeneration. Cells 2022; 11:cells11030397. [PMID: 35159206 PMCID: PMC8834155 DOI: 10.3390/cells11030397] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/27/2023] Open
Abstract
Sterile inflammation either resolves the initial insult or leads to tissue damage. Kidney ischemia/reperfusion injury (IRI) is associated with neutrophilic infiltration, enhanced production of inflammatory mediators, accumulation of necrotic cells and tissue remodeling. Macrophage-dependent microenvironmental changes orchestrate many features of the immune response and tissue regeneration. The activation status of macrophages is influenced by extracellular signals, the duration and intensity of the stimulation, as well as various regulatory molecules. The role of macrophage-derived monocyte chemoattractant protein-induced protein 1 (MCPIP1), also known as Regnase-1, in kidney ischemia-reperfusion injury (IRI) and recovery from sterile inflammation remains unresolved. In this study, we showed that macrophage-specific Mcpip1 deletion significantly affects the kidney phenotype. Macrophage-specific Mcpip1 transgenic mice displayed enhanced inflammation and loss of the tubular compartment upon IRI. We showed that MCPIP1 modulates sterile inflammation by negative regulation of Irf4 expression and accumulation of IRF4+ cells in the tissue and, consequently, suppresses the post-ischemic kidney immune response. Thus, we identified MCPIP1 as an important molecular sentinel of immune homeostasis in experimental acute kidney injury (AKI) and renal fibrosis.
Collapse
Affiliation(s)
- Andrea Ribeiro
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
- Department of Nephrology, Klinikum Rechts der Isar, Technical University Munich, 80336 Munich, Germany;
| | - Ewelina Dobosz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (E.D.); (M.W.); (J.K.)
| | - Moritz Krill
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
| | - Paulina Köhler
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
| | - Marta Wadowska
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (E.D.); (M.W.); (J.K.)
| | - Stefanie Steiger
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum Rechts der Isar, Technical University Munich, 80336 Munich, Germany;
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (E.D.); (M.W.); (J.K.)
| | - Maciej Lech
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
- Correspondence:
| |
Collapse
|
4
|
Jawad A, Yoo YJ, Yoon JC, Tian W, Islam MS, Lee EY, Shin HY, Kim SE, Ahn D, Park BY, Tae HJ, Kim IS. Changes of renal histopathology and the role of Nrf2/HO-1 in asphyxial cardiac arrest model in rats. Acta Cir Bras 2021; 36:e360607. [PMID: 34287609 PMCID: PMC8291904 DOI: 10.1590/acb360607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/12/2021] [Indexed: 01/17/2023] Open
Abstract
PURPOSE To investigate the role of Nrf2/HO-1 in renal histopathological ailments time-dependently in asphyxial cardiac arrest (CA) rat model. METHODS Eighty-eight Sprague Dawley male rats were divided into five groups of eight rats each. Asphyxial CA was induced in all the experimental rats except for the sham group. The rats were sacrificed at 6 hours, 12 hours, one day and two days post-CA. Serum blood urea nitrogen (BUN), creatinine (Crtn) and malondialdehyde from the renal tissues were evaluated. Hematoxylin and eosin and periodic acid-Schiff staining were done to evaluate the renal histopathological changes in the renal cortex. Furthermore, Nrf2/HO-1 immunohistochemistry (ihc) and western blot analysis were performed after CA. RESULTS The survival rate of rats decreased in a time-dependent manner: 66.6% at 6 hours, 50% at 12 hours, 38.1% in one day, and 25.8% in two days. BUN and serum Crtn markedly increased in CA-operated groups. Histopathological ailments of the renal cortical tissues increased significantly from 6 hours until two days post-CA. Furthermore, Nrf2/HO-1 expression level significantly increased at 6 hours, 12 hours, and one day. CONCLUSIONS The survival rate decreased time-dependently, and Nrf/HO-1 expression increased from 6 hours with the peak times at 12 hours, and one day post-CA.
Collapse
Affiliation(s)
- Ali Jawad
- Jeonbuk National University, South Korea
| | | | | | | | | | | | | | - So Eun Kim
- Jeonbuk National University Hospital, South Korea
| | | | | | | | | |
Collapse
|
5
|
Flacourtia indica fruit extract modulated antioxidant gene expression, prevented oxidative stress and ameliorated kidney dysfunction in isoprenaline administered rats. Biochem Biophys Rep 2021; 26:101012. [PMID: 34041370 PMCID: PMC8142055 DOI: 10.1016/j.bbrep.2021.101012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/13/2021] [Accepted: 04/30/2021] [Indexed: 11/22/2022] Open
Abstract
This study evaluated the effect of Flacourtia indica fruit extract against isoprenaline (ISO) induced renal damage in rats. This investigation showed that ISO administration in rats increased the level oxidative stress biomarkers such as malondialdehyde (MDA), nitric oxide (NO), advanced protein oxidation product (APOP) in kidneys followed by a decrease in antioxidant enzymes functions. Flacourtia indica fruit extract, which is rich in strong antioxidants, also reduced the MDA, NO and APOP level in kidney of ISO administered rats. Inflammation and necrosis was also visible in kidney section of ISO administered rats which was significantly prevented by atenolol and Flacourtia indica fruit extract. Moreover, atenolol and Flacourtia indica fruit extract also modulated the genes expressions related to inflammation and oxidative stress in kidneys. The beneficial effects could be attributed to the presence of a number of phenolic antioxidants. This study suggests that Flacourtia indica fruit extract may prevent kidney dysfunction in ISO administered rats, probably by preventing oxidative stress and inflammation.
Collapse
|
6
|
Long-term Efficacy and Safety of Everolimus Versus Mycophenolate in Kidney Transplant Recipients Receiving Tacrolimus. Transplantation 2021; 106:381-390. [PMID: 33988338 DOI: 10.1097/tp.0000000000003714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The short-term efficacy and safety of everolimus in combination with tacrolimus have been described in several clinical trials. Yet, detailed long-term data comparing the use of everolimus or mycophenolate in kidney transplant recipients receiving tacrolimus is lacking. METHODS This is a 5-year follow-up post hoc analysis of a prospective trial including 288 patients who were randomized to receive a single 3 mg/kg dose of rabbit antithymocyte globulin, tacrolimus, everolimus, and prednisone (r-ATG/EVR, n=85); basiliximab, tacrolimus, everolimus, and prednisone (BAS/EVR, n=102); or basiliximab, tacrolimus, mycophenolate, and prednisone (BAS/MPS, n=101). RESULTS There were no differences in the incidence of treatment failure (31.8% vs. 40.2% vs. 34.7%, p=0.468), de novo donor-specific HLA antibodies (6.5 vs. 11.7 vs. 4.0%, p=0.185), patient (92.9% vs. 94.1% vs. 92.1%, p = 0.854) and death-censored graft (87.1% vs. 90.2% vs. 85.1%, p = 0.498) survivals. Using a sensitive analysis, the trajectories of eGFR were comparable in the intention-to-treat (p=0.145) and per protocol (p=0.354) populations. There were no differences in study drug discontinuation rate (22.4% vs. 30.4% vs. 17.8%, p=0.103). CONCLUSIONS In summary, this analysis in a cohort of de novo low/moderate immunologic risk kidney transplant recipients suggests that the use of a single 3mg/kg r-ATG dose followed by EVR combined with reduced TAC concentrations was associated with similar efficacy and renal function compared to the standard of care immunosuppressive regimen.Supplemental Visual Abstract; http://links.lww.com/TP/C160.
Collapse
|
7
|
Grunenwald A, Roumenina LT, Frimat M. Heme Oxygenase 1: A Defensive Mediator in Kidney Diseases. Int J Mol Sci 2021; 22:2009. [PMID: 33670516 PMCID: PMC7923026 DOI: 10.3390/ijms22042009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
The incidence of kidney disease is rising, constituting a significant burden on the healthcare system and making identification of new therapeutic targets increasingly urgent. The heme oxygenase (HO) system performs an important function in the regulation of oxidative stress and inflammation and, via these mechanisms, is thought to play a role in the prevention of non-specific injuries following acute renal failure or resulting from chronic kidney disease. The expression of HO-1 is strongly inducible by a wide range of stimuli in the kidney, consequent to the kidney's filtration role which means HO-1 is exposed to a wide range of endogenous and exogenous molecules, and it has been shown to be protective in a variety of nephropathological animal models. Interestingly, the positive effect of HO-1 occurs in both hemolysis- and rhabdomyolysis-dominated diseases, where the kidney is extensively exposed to heme (a major HO-1 inducer), as well as in non-heme-dependent diseases such as hypertension, diabetic nephropathy or progression to end-stage renal disease. This highlights the complexity of HO-1's functions, which is also illustrated by the fact that, despite the abundance of preclinical data, no drug targeting HO-1 has so far been translated into clinical use. The objective of this review is to assess current knowledge relating HO-1's role in the kidney and its potential interest as a nephroprotection agent. The potential therapeutic openings will be presented, in particular through the identification of clinical trials targeting this enzyme or its products.
Collapse
Affiliation(s)
- Anne Grunenwald
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Lubka T. Roumenina
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Marie Frimat
- U1167-RID-AGE, Institut Pasteur de Lille, Inserm, Univ. Lille, F-59000 Lille, France
- Nephrology Department, CHU Lille, Univ. Lille, F-59000 Lille, France
| |
Collapse
|
8
|
Hyperbaric Oxygen Preconditioning Upregulates Heme OxyGenase-1 and Anti-Apoptotic Bcl-2 Protein Expression in Spontaneously Hypertensive Rats with Induced Postischemic Acute Kidney Injury. Int J Mol Sci 2021; 22:ijms22031382. [PMID: 33573145 PMCID: PMC7866496 DOI: 10.3390/ijms22031382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/19/2022] Open
Abstract
Renal ischemia and reperfusion (I/R) injury is the most common cause of acute kidney injury (AKI). Pathogenesis of postischemic AKI involves hemodynamic changes, oxidative stress, inflammation process, calcium ion overloading, apoptosis and necrosis. Up to date, therapeutic approaches to treat AKI are extremely limited. Thus, the aim of this study was to evaluate the effects of hyperbaric oxygen (HBO) preconditioning on citoprotective enzyme, heme oxygenase-1 (HO-1), pro-apoptotic Bax and anti-apoptotic Bcl-2 proteins expression, in postischemic AKI induced in normotensive Wistar and spontaneously hypertensive rats (SHR). The animals were randomly divided into six experimental groups: SHAM-operated Wistar rats (W-SHAM), Wistar rats with induced postischemic AKI (W-AKI) and Wistar group with HBO preconditioning before AKI induction (W-AKI + HBO). On the other hand, SHR rats were also divided into same three groups: SHR-SHAM, SHR-AKI and SHR-AKI + HBO. We demonstrated that HBO preconditioning upregulated HO-1 and anti-apoptotic Bcl-2 protein expression, in both Wistar and SH rats. In addition, HBO preconditioning improved glomerular filtration rate, supporting by significant increase in creatinine, urea and phosphate clearances in both rat strains. Considering our results, we can also say that even in hypertensive conditions, we can expect protective effects of HBO preconditioning in experimental model of AKI.
Collapse
|
9
|
Fu R, Tajima S, Shigematsu T, Zhang M, Tsuchimoto A, Egashira N, Ieiri I, Masuda S. Establishment of an experimental rat model of tacrolimus-induced kidney injury accompanied by interstitial fibrosis. Toxicol Lett 2021; 341:43-50. [PMID: 33516819 DOI: 10.1016/j.toxlet.2021.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
Nephrotoxicity is the major adverse reaction to tacrolimus; however, the underlying mechanisms remain to be fully elucidated. Although several tacrolimus-induced nephrotoxicity animal models have been reported, most renal injury rat models contain factors other than tacrolimus. Here, we report the development of a new nephrotoxicity with interstitial fibrosis rat model induced by tacrolimus administration. Thirty Wistar rats were randomly divided into four groups: sham-operated (Sham), vehicle-treated ischemia reperfusion (I/R) injury (IRI), tacrolimus treated (TAC) and tacrolimus treated I/R injury (TAC + IRI). Rats subjected to IR injury and treated with tacrolimus for 2 weeks showed higher serum creatinine (Scr), blood urea nitrogen (BUN), serum magnesium (Mg) and serum potassium (K), indicating decreased renal function. In addition, tacrolimus treatment combined with IR injury increased histological injury (tubular vacuolation, glomerulosclerosis and interstitial fibrosis), as well as α-smooth muscle actin (α-SMA), transforming growth factor-β (TGF-β), and kidney injury molecule-1 (KIM-1) expression in the renal cortex. In summary, we have developed a tacrolimus-induced kidney injury rat model with interstitial fibrosis within 2 weeks by creating conditions mimicking renal transplantation via tacrolimus administration following ischemia-reperfusion.
Collapse
Affiliation(s)
- Rao Fu
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Japan
| | | | - Tomohiro Shigematsu
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Japan; Department of Pharmacy, Kyushu University Hospital, Japan
| | - Mengyu Zhang
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Japan
| | - Akihiro Tsuchimoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Nobuaki Egashira
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Japan; Department of Pharmacy, Kyushu University Hospital, Japan
| | - Ichiro Ieiri
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Japan; Department of Pharmacy, Kyushu University Hospital, Japan
| | - Satohiro Masuda
- Department of Pharmacy, International University of Health and Welfare Narita Hospital, Japan; Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, International University of Health and Welfare, Japan
| |
Collapse
|
10
|
Abbasi Dezfouli S, Nikdad M, Ghamarnejad O, Khajeh E, Arefidoust A, Mohammadi S, Majlesara A, Sabagh M, Gharabaghi N, Kentar M, Younsi A, Eckert C, Poth T, Golriz M, Mehrabi A, Nickkholgh A. Oral Preconditioning of Donors After Brain Death With Calcineurin Inhibitors vs. Inhibitors of Mammalian Target for Rapamycin in Pig Kidney Transplantation. Front Immunol 2020; 11:1222. [PMID: 32625210 PMCID: PMC7316124 DOI: 10.3389/fimmu.2020.01222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/15/2020] [Indexed: 12/01/2022] Open
Abstract
Background: The systemic inflammatory cascade triggered in donors after brain death enhances the ischemia-reperfusion injury after organ transplantation. Intravenous steroids are routinely used in the intensive care units for the donor preconditioning. Immunosuppressive medications could be potentially used for this purpose as well. Data regarding donor preconditioning with calcineurin inhibitors or inhibitors of mammalian target for Rapamycin is limited. The aim of this project is to investigate the effects of (oral) donor preconditioning with a calcineurin inhibitor (Cyclosporine) vs. an inhibitor of mammalian target for Rapamycin (Everolimus) compared to the conventional administration of steroid in the setting of donation after brain death in porcine renal transplantation. Methods: Six hours after the induction of brain death, German landrace donor pigs (33.2 ± 3.9 kg) were randomly preconditioned with either Cyclosporine (n = 9) or Everolimus (n = 9) administered via nasogastric tube with a repeated dose just before organ procurement. Control donors received intravenous Methylprednisolone (n = 8). Kidneys were procured, cold-stored in Histidine-Tryptophane-Ketoglutarate solution at 4°C and transplanted in nephrectomized recipients after a mean cold ischemia time of 18 h. No post-transplant immunosuppression was given to avoid confounding bias. Blood samples were obtained at 4 h post reperfusion and daily until postoperative day 5 for complete blood count, blood urea nitrogen, creatinine, and electrolytes. Graft protocol biopsies were performed 4 h after reperfusion to assess early histological and immunohistochemical changes. Results: There was no difference in the hemodynamic parameters, hemoglobin/hematocrit and electrolytes between the groups. Serum blood urea nitrogen and creatinine peaked on postoperative day 1 in all groups and went back to the preoperative levels at the conclusion of the study on postoperative day 5. Histological assessment of the kidney grafts revealed no significant differences between the groups. TNF-α expression was significantly lower in the study groups compared with Methylprednisolone group (p = 0.01) Immunohistochemistry staining for cytochrome c showed no difference between the groups. Conclusion: Oral preconditioning with Cyclosporine or Everolimus is feasible in donation after brain death pig kidney transplantation and reduces the expression of TNF-α. Future studies are needed to further delineate the role of oral donor preconditioning against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Sepehr Abbasi Dezfouli
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Mohammadsadegh Nikdad
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Omid Ghamarnejad
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Elias Khajeh
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Alireza Arefidoust
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Sara Mohammadi
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Ali Majlesara
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Mohammadsadegh Sabagh
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Negin Gharabaghi
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Modar Kentar
- Department of Neurosurgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Christoph Eckert
- Institute of Pathology, Ruprecht-Karls University, Heidelberg, Germany
| | - Tanja Poth
- Institute of Pathology, Ruprecht-Karls University, Heidelberg, Germany
| | - Mohammad Golriz
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Arianeb Mehrabi
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| | - Arash Nickkholgh
- Department of General, Abdominal and Transplant Surgery, Ruprecht-Karls University, Heidelberg, Germany
| |
Collapse
|
11
|
Corona D, Ekser B, Gioco R, Caruso M, Schipa C, Veroux P, Giaquinta A, Granata A, Veroux M. Heme-Oxygenase and Kidney Transplantation: A Potential for Target Therapy? Biomolecules 2020; 10:E840. [PMID: 32486245 PMCID: PMC7355572 DOI: 10.3390/biom10060840] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/24/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022] Open
Abstract
Kidney transplantation is a well-established therapy for patients with end-stage renal disease. While a significant improvement of short-term results has been achieved in the short-term, similar results were not reported in the long-term. Heme-oxygenase (HO) is the rate-limiting enzyme in heme catabolism, converting heme to iron, carbon monoxide, and biliverdin. Heme-oxygenase overexpression may be observed in all phases of transplant processes, including brain death, recipient management, and acute and chronic rejection. HO induction has been proved to provide a significant reduction of inflammatory response and a reduction of ischemia and reperfusion injury in organ transplantation, as well as providing a reduction of incidence of acute rejection. In this review, we will summarize data on HO and kidney transplantation, suggesting possible clinical applications in the near future to improve the long-term outcomes.
Collapse
Affiliation(s)
- Daniela Corona
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (D.C.); (M.C.)
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
| | - Burcin Ekser
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46077, USA;
| | - Rossella Gioco
- General Surgery Unit, University Hospital of Catania, 95123 Catania, Italy; (R.G.); (C.S.)
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (D.C.); (M.C.)
| | - Chiara Schipa
- General Surgery Unit, University Hospital of Catania, 95123 Catania, Italy; (R.G.); (C.S.)
| | - Pierfrancesco Veroux
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
| | - Alessia Giaquinta
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
| | | | - Massimiliano Veroux
- Organ Transplant Unit, University Hospital of Catania, 95123 Catania, Italy; (P.V.); (A.G.)
- General Surgery Unit, University Hospital of Catania, 95123 Catania, Italy; (R.G.); (C.S.)
| |
Collapse
|
12
|
Kim DJ, Moon JY, Kim SM, Seo JW, Lee YH, Jung SW, Kim K, Kim YG, Lim SJ, Lee S, Son Y, Lee SH. Substance P Improves Renal Ischemia Reperfusion Injury Through Modulating Immune Response. Front Immunol 2020; 11:600. [PMID: 32391002 PMCID: PMC7190869 DOI: 10.3389/fimmu.2020.00600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/16/2020] [Indexed: 12/28/2022] Open
Abstract
Substance P (SP), an injury-inducible messenger that mobilizes bone marrow stem cells and modulates the immune response, has been suggested as a novel target for therapeutic agents. We evaluated the role of SP as an immune cell modulator during the progression of renal ischemic/reperfusion injury (IRI). Unilateral IRI induced the transient expression of endogenous SP and the infiltration of CCR7+ M1 macrophages in injured kidneys. However, SP altered the intrarenal macrophage polarization from CCR7+ M1 macrophages to CD206+ M2 macrophages in injured kidneys. SP also modulated bone marrow-derived neutrophils and mesenchymal stromal cells after IRI. SP treatment for 4 weeks starting one week after unilateral IRI significantly preserved kidney size and length and normal tubular structures and alleviated necrotic tubules, inflammation, apoptosis, and tubulointerstitial fibrosis. The beneficial effects of SP were accompanied by attenuation of intrarenal recruitment of CD4, CD8, and CD20 cells and abnormal angiogenesis. The immunomodulatory effect of SP suggested that SP could be a promising therapeutic target for preventing the progression of acute kidney injury to chronic kidney disease.
Collapse
Affiliation(s)
- Dong-Jin Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea.,Laboratory of Tissue Engineering, Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University Global Campus, Yongin, South Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Su-Mi Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Jung-Woo Seo
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Yu Ho Lee
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Kipyo Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, College of Medicine, Inha University, Incheon, South Korea
| | - Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Sung-Jig Lim
- Department of Pathology, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | | | - Youngsook Son
- Laboratory of Tissue Engineering, Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University Global Campus, Yongin, South Korea
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
13
|
Rund KM, Peng S, Greite R, Claaßen C, Nolte F, Oger C, Galano JM, Balas L, Durand T, Chen R, Gueler F, Schebb NH. Dietary omega-3 PUFA improved tubular function after ischemia induced acute kidney injury in mice but did not attenuate impairment of renal function. Prostaglandins Other Lipid Mediat 2019; 146:106386. [PMID: 31698142 DOI: 10.1016/j.prostaglandins.2019.106386] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/09/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is an important complication after major surgery and solid organ transplantation. Here, we present a dietary omega-3 polyunsaturated fatty acid (n3-PUFA) supplementation study to investigate whether pre-treatment can reduce ischemia induced AKI in mice. METHODS Male 12-14 week old C57BL/6 J mice received a linoleic acid rich sunflower oil based standard diet containing 10 % fat (STD) or the same diet enriched with n3-PUFA (containing 1 % EPA and 1 % DHA) (STD + n3). After 14 days of feeding bilateral 30 min renal ischemia reperfusion injury (IRI) was conducted to induce AKI and mice were sacrificed at 24 h. Serum creatinine and blood urea nitrogen (BUN) as well as liver enzyme elevation were measured. Kidney damage was analyzed by histology and immunohistochemistry. Furthermore, pro-inflammatory cytokines (IL-6, MCP-1) were determined by qPCR. FA and oxylipin pattern were quantified in blood and kidneys by GC-FID and LC-MS/MS, respectively. RESULTS n3-PUFA supplementation prior to renal IRI increased systemic and renal levels of n3-PUFA. Consistently, eicosanoids and other oxylipins derived from n3-PUFA including precursors of specialized pro-resolving mediators were elevated while n6-PUFA derived mediators such as pro-inflammatory prostaglandins were decreased. Feeding of n3-PUFA did not attenuate renal function impairment, morphological renal damage and inflammation characterized by IL-6 and MCP-1 elevation or neutrophil infiltration. However, the tubular transport marker alpha-1 microglobulin (A1M) was significantly higher expressed in proximal tubular epithelial cells of STD + n3 compared to STD fed mice. This indicates a better integrity of proximal tubular epithelial cells and thus significant protection of tubular function. In addition, heme oxygenase-1 (HO-1) which protects tubular function was also up-regulated in the treatment group receiving n3-PUFA supplemented chow. DISCUSSION We showed that n3-PUFA pre-treatment did not affect overall renal function or renal inflammation in a mouse model of moderate ischemia induced AKI, but tubular transport was improved. In conclusion, dietary n3-PUFA supplementation altered the oxylipin levels significantly but did not protect from renal function deterioration or attenuate ischemia induced renal inflammation.
Collapse
Affiliation(s)
- Katharina M Rund
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Shu Peng
- Nephrology, Hannover Medical School, Hannover, Germany; Department of Thoracic surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Robert Greite
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Cornelius Claaßen
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Fabian Nolte
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Laurence Balas
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Rongjun Chen
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Faikah Gueler
- Nephrology, Hannover Medical School, Hannover, Germany.
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany.
| |
Collapse
|
14
|
Bottino-Rojas V, Pereira LOR, Silva G, Talyuli OAC, Dunkov BC, Oliveira PL, Paiva-Silva GO. Non-canonical transcriptional regulation of heme oxygenase in Aedes aegypti. Sci Rep 2019; 9:13726. [PMID: 31551499 PMCID: PMC6760526 DOI: 10.1038/s41598-019-49396-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenase (HO) is a ubiquitous enzyme responsible for heme breakdown, which yields carbon monoxide (CO), biliverdin (BV) and ferrous ion. Here we show that the Aedes aegypti heme oxygenase gene (AeHO - AAEL008136) is expressed in different developmental stages and tissues. AeHO expression increases after a blood meal in the midgut, and its maximal transcription levels overlaps with the maximal rate of the further modified A. aegypti biglutaminyl-biliverdin (AeBV) pigment production. HO is a classical component of stress response in eukaryotic cells, being activated under oxidative stress or increased heme levels. Indeed, the final product of HO activity in the mosquito midgut, AeBV, exerts a protective antioxidant activity. AeHO, however, does not seem to be under a classical redox-sensitive transcriptional regulation, being unresponsive to heme itself, and even down regulated when insects face a pro-oxidant insult. In contrast, AeHO gene expression responds to nutrient sensing mechanisms, through the target of rapamycin (TOR) pathway. This unusual transcriptional control of AeHO, together with the antioxidant properties of AeBV, suggests that heme degradation by HO, in addition to its important role in protection of Aedes aegypti against heme exposure, also acts as a digestive feature, being an essential adaptation to blood feeding.
Collapse
Affiliation(s)
- Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Luiza O R Pereira
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, 21040-360, Brazil
| | - Gabriela Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Boris C Dunkov
- Center for Insect Science, The University of Arizona, Tucson, AZ, 85721-0106, USA
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
15
|
Li S, Qiu B, Lu H, Lai Y, Liu J, Luo J, Zhu F, Hu Z, Zhou M, Tian J, Zhou Z, Yu S, Yi F, Nie J. Hyperhomocysteinemia Accelerates Acute Kidney Injury to Chronic Kidney Disease Progression by Downregulating Heme Oxygenase-1 Expression. Antioxid Redox Signal 2019; 30:1635-1650. [PMID: 30084650 DOI: 10.1089/ars.2017.7397] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIMS The risk factors promoting acute kidney injury (AKI) to chronic kidney disease (CKD) progression remain largely unknown. The aim of the present study was to investigate whether hyperhomocysteinemia (Hhcy) accelerates the development of renal fibrosis after AKI. RESULTS Hhcy aggravated ischemia-reperfusion-induced AKI and the subsequent development of renal fibrotic lesions characterized by excessive extracellular matrix deposition. Mechanistically, the RNA binding protein human antigen R (HuR) bound to the 3'-untranslated region (3'-UTR) of heme oxygenase-1 (HO-1) messenger RNA (mRNA). Homocysteine (Hcy) downregulated HuR expression, reduced the binding of HuR to the 3'-UTR of HO-1, and thereafter decreased HO-1 expression. Administration of the HO-1 inducer cobalt protoporphyrin-IX significantly hindered Hhcy-augmented reactive oxygen species production and renal fibrotic lesions. Innovation and Conclusion: These data indicate that Hhcy might be a novel risk factor that promotes AKI to CKD progression. Lowering Hcy level or HO-1 induction might be a potential therapeutic strategy to improve the outcome of AKI.
Collapse
Affiliation(s)
- Shuang Li
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingbing Qiu
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong Lu
- 2 Department of Public Health, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yunshi Lai
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jixing Liu
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiajun Luo
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fengxin Zhu
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zheng Hu
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Miaomiao Zhou
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianwei Tian
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhanmei Zhou
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shouyi Yu
- 2 Department of Public Health, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fan Yi
- 3 Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Jing Nie
- 1 State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Kim JE, Bae SY, Ahn SY, Kwon YJ, Ko GJ. The role of nuclear factor erythroid-2-related factor 2 expression in radiocontrast-induced nephropathy. Sci Rep 2019; 9:2608. [PMID: 30796317 PMCID: PMC6384919 DOI: 10.1038/s41598-019-39534-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 01/10/2019] [Indexed: 01/21/2023] Open
Abstract
Radiocontrast-induced nephropathy (CIN) is the third most common cause of acute renal failure. The pathophysiology of CIN is related to tubular injury caused by oxidative stress, and nuclear factor erythroid-2-related factor 2 (Nrf2) is critical in coordinating intracellular antioxidative processes. We thus investigated the role of Nrf2 in CIN. CIN was established in mice and in NRK-52E cells via iohexol administration according to the protocols of previous studies. To determine the role of Nrf2 in CIN, Nrf2 expression was reduced in vivo using Nrf2 knockout (KO) mice (B6.129 × 1-Nfe2 l2tm1Ywk/J) and in vitro with siRNA treatment targeting Nrf2. Increased Nrf2 expression was observed after iohexol treatment both in vivo and in vitro. Serum creatinine at 24 h after iohexol injection was significantly higher in KO mice than in wild-type (WT) mice. Histologic examination showed that iohexol-induced tubular vacuolization and structural disruption were aggravated in Nrf2 KO mice. Significant increases in apoptosis and F4/80(+) inflammatory cell infiltration were demonstrated in KO mice compared to WT mice. In addition, the increase in reactive oxygen species after iohexol treatment was augmented by Nrf2 inhibition both in vivo and in vitro. Nrf2 may be implicated in the pathogenesis of CIN via the modulation of antioxidant, anti-apoptotic, and anti-inflammatory processes.
Collapse
Affiliation(s)
- Ji Eun Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - So Yeon Bae
- Nephrology Research Institution, Korea University Guro Hospital, Seoul, Korea
| | - Shin Young Ahn
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Young Joo Kwon
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Gang Jee Ko
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
17
|
Liu D, Xu L, Zhang X, Shi C, Qiao S, Ma Z, Yuan J. Snapshot: Implications for mTOR in Aging-related Ischemia/Reperfusion Injury. Aging Dis 2019; 10:116-133. [PMID: 30705773 PMCID: PMC6345330 DOI: 10.14336/ad.2018.0501] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 05/01/2018] [Indexed: 12/15/2022] Open
Abstract
Aging may aggravate the damage and dysfunction of different components of multiorgan and thus increasing multiorgan ischemia/reperfusion (IR) injury. IR injury occurs in many organs and tissues, which is a major cause of morbidity and mortality worldwide. The kinase mammalian target of rapamycin (mTOR), an atypical serine/threonine protein kinase, involves in the pathophysiological process of IR injury. In this review, we first briefly introduce the molecular features of mTOR, the association between mTOR and aging, and especially its role on autophagy. Special focus is placed on the roles of mTOR during ischemic and IR injury. We then clarify the association between mTOR and conditioning phenomena. Following this background, we expand our discussion to potential future directions of research in this area. Collectively, information reviewed herein will serve as a comprehensive reference for the actions of mTOR in IR injury and may be significant for the design of future research and increase the potential of mTOR as a therapeutic target.
Collapse
Affiliation(s)
- Dong Liu
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Liqun Xu
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China.,3Cadet group 3, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China.,4Laboratory Animal Center, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoyan Zhang
- 2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China.,3Cadet group 3, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China
| | - Changhong Shi
- 4Laboratory Animal Center, The Fourth Military Medical University, Xi'an 710032, China
| | - Shubin Qiao
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhiqiang Ma
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Jiansong Yuan
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| |
Collapse
|
18
|
Jankauskas SS, Silachev DN, Andrianova NV, Pevzner IB, Zorova LD, Popkov VA, Plotnikov EY, Zorov DB. Aged kidney: can we protect it? Autophagy, mitochondria and mechanisms of ischemic preconditioning. Cell Cycle 2018; 17:1291-1309. [PMID: 29963970 DOI: 10.1080/15384101.2018.1482149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The anti-aging strategy is one of the main challenges of the modern biomedical science. The term "aging" covers organisms, cells, cellular organelles and their constituents. In general term, aging system admits the existence of nonfunctional structures which by some reasons have not been removed by a clearing system, e.g., through autophagy/mitophagy marking and destroying unwanted cells or mitochondria. This directly relates to the old kidney which normal functioning is critical for the viability of the organism. One of the main problems in biomedical studies is that in their majority, young organisms serve as a standard with further extrapolation on the aged system. However, some protective systems, which demonstrate their efficiency in young systems, lose their beneficial effect in aged organisms. It is true for ischemic preconditioning of the kidney, which is almost useless for an old kidney. The pharmacological intervention could correct the defects of the senile system provided that the complete understanding of all elements involved in aging will be achieved. We discuss critical elements which determine the difference between young and old phenotypes and give directions to prevent or cure lesions occurring in aged organs including kidney. ABBREVIATIONS AKI: acute kidney injury; I/R: ischemia/reperfusion; CR: caloric restriction; ROS: reactive oxygen species; RC: respiratory chain.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation
| | - Denis N Silachev
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,b Department of Molecular Mechanisms of Adaptation , V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology , Moscow , Russian Federation
| | - Nadezda V Andrianova
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,c Faculty of Bioengineering and Bioinformatics , M.V. Lomonosov Moscow State University , Moscow , Russian Federation
| | - Irina B Pevzner
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,b Department of Molecular Mechanisms of Adaptation , V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology , Moscow , Russian Federation
| | - Ljubava D Zorova
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,b Department of Molecular Mechanisms of Adaptation , V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology , Moscow , Russian Federation
| | - Vasily A Popkov
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,c Faculty of Bioengineering and Bioinformatics , M.V. Lomonosov Moscow State University , Moscow , Russian Federation
| | - Egor Y Plotnikov
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,b Department of Molecular Mechanisms of Adaptation , V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology , Moscow , Russian Federation
| | - Dmitry B Zorov
- a A.N. Belozersky Institute of Physico-Chemical Biology , M.V. Lomonosov Moscow State University , Moscow , Russian Federation.,b Department of Molecular Mechanisms of Adaptation , V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology , Moscow , Russian Federation
| |
Collapse
|
19
|
Roles of mTOR complexes in the kidney: implications for renal disease and transplantation. Nat Rev Nephrol 2016; 12:587-609. [PMID: 27477490 DOI: 10.1038/nrneph.2016.108] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR pathway has a central role in the regulation of cell metabolism, growth and proliferation. Studies involving selective gene targeting of mTOR complexes (mTORC1 and mTORC2) in renal cell populations and/or pharmacologic mTOR inhibition have revealed important roles of mTOR in podocyte homeostasis and tubular transport. Important advances have also been made in understanding the role of mTOR in renal injury, polycystic kidney disease and glomerular diseases, including diabetic nephropathy. Novel insights into the roles of mTORC1 and mTORC2 in the regulation of immune cell homeostasis and function are helping to improve understanding of the complex effects of mTOR targeting on immune responses, including those that impact both de novo renal disease and renal allograft outcomes. Extensive experience in clinical renal transplantation has resulted in successful conversion of patients from calcineurin inhibitors to mTOR inhibitors at various times post-transplantation, with excellent long-term graft function. Widespread use of this practice has, however, been limited owing to mTOR-inhibitor- related toxicities. Unique attributes of mTOR inhibitors include reduced rates of squamous cell carcinoma and cytomegalovirus infection compared to other regimens. As understanding of the mechanisms by which mTORC1 and mTORC2 drive the pathogenesis of renal disease progresses, clinical studies of mTOR pathway targeting will enable testing of evolving hypotheses.
Collapse
|
20
|
Zhao S, Wei Y, Xu D. Neutrophil gelatinase-associated lipocalin attenuates injury in the rat cecal ligation and puncture model of sepsis via apoptosis inhibition. Nephrology (Carlton) 2016; 20:646-53. [PMID: 25943501 DOI: 10.1111/nep.12498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 11/28/2022]
Abstract
AIM The aim of this study was to investigate the effect of neutrophil gelatinase-associated lipocalin (NGAL) on the rat cecal ligation and puncture (CLP)-induced sepsis and the possible mechanism. METHODS Thirty male Sprague-Dawley rats underwent CLP as sepsis models and were randomized into three groups including the sham-operated group (sham, n = 10), which only underwent a laparotomy; the sepsis group (sepsis, n = 10), which underwent CLP and subcutaneous injection of normal saline; and the sepsis + NGAL group (sepsis + NGAL, n = 10), which underwent CLP and subcutaneous injection of NGAL. Urine, blood and kidney tissue samples were collected for the determination of urine NGAL (uNGAL), plasma NGAL (pNGAL), serum creatinine (Scr), blood urea nitrogen (BUN), histomorphological and immunohistochemical examination, lipid peroxidation product malondialdehyde (MDA) and superoxide dismutase (SOD), and expression of heme oxygenase-1 (HO)-1. RESULTS The levels of uNGAL, pNGAL, Scr, BUN, kidney injury score, positive TUNEL staining, activated Caspase-3 and Bax, and kidney tissue MDA levels in the sepsis group were significantly increased compared with those in the sham-operated group and the sepsis + NGAL group (P < 0.05). SOD level and HO-1 expression in sepsis + NGAL group were significantly higher than those in the sham-operated group and the sepsis group (P < 0.05). CONCLUSION NGAL can attenuate kidney injury and apoptosis in the rat CLP model of sepsis. And the protective effect of NGAL was probably due to the inhibition of apoptosis and lipid peroxidation, and increased expression of HO-1.
Collapse
Affiliation(s)
- Shuangping Zhao
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yangjing Wei
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daomiao Xu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Chen HH, Lu PJ, Chen BR, Hsiao M, Ho WY, Tseng CJ. Heme oxygenase-1 ameliorates kidney ischemia-reperfusion injury in mice through extracellular signal-regulated kinase 1/2-enhanced tubular epithelium proliferation. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2195-201. [PMID: 26232688 DOI: 10.1016/j.bbadis.2015.07.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 01/01/2023]
Abstract
Heme oxygenase (HO)-1 confers transient resistance against oxidative damage, including renal ischemia-reperfusion injury (IRI). We investigated the potential protective effect of HO-1 induction in a mouse model of renal IRI induced by bilateral clamping of the kidney arteries. The mice were randomly assigned to five groups to receive an intraperitoneal injection of PBS, hemin (an HO-1 inducer, 100μmol/kg), hemin+ZnPP (an HO-1 inhibitor, 5mg/kg), hemin+PD98059 (a MEK-ERK inhibitor, 10mg/kg) or a sham operation. All of the groups except for the sham-operated group underwent 25min of ischemia and 24 to 72h of reperfusion. Renal function and tubular damage were assessed in the mice that received hemin or the vehicle treatment prior to IRI. The renal injury score and HO-1 protein levels were evaluated via H&E and immunohistochemistry staining. Hemin-preconditioned mice exhibited preserved renal cell function (BUN: 40±2mg/dl, creatinine: 0.53±0.06mg/dl), and the tubular injury score at 72h (1.65±0.12) indicated that tubular damage was prevented. Induction of HO-1 induced the phosphorylation of extracellular signal-regulated kinases (ERK) 1/2. However, these effects were abolished with ZnPP treatment. Kidney function (BUN: 176±49mg/dl, creatinine: 1.54±0.39mg/dl) increased, and the tubular injury score (3.73±0.09) indicated that tubular damage also increased with ZnPP treatment. HO-1-induced tubular epithelial proliferation was attenuated by PD98059. Our findings suggest that HO-1 preconditioning promotes ERK1/2 phosphorylation and enhances tubular recovery, which subsequently prevents further renal injury.
Collapse
Affiliation(s)
- Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Yuh-Ing Junior College of Health Care & Management, Kaohsiung, Taiwan
| | - Pei-Jung Lu
- Graduate Institute of Clinical Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Bo-Ron Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Yu Ho
- Division of General Internal Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Internal Medicine, Faculty of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Ching-Jiunn Tseng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
22
|
Rapamycin protection of livers from ischemia and reperfusion injury is dependent on both autophagy induction and mammalian target of rapamycin complex 2-Akt activation. Transplantation 2015; 99:48-55. [PMID: 25340604 DOI: 10.1097/tp.0000000000000476] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Although rapamycin (RPM) have been studied extensively in ischemia models, its functional mechanisms remains to be defined. METHODS We determined how RPM impacted the pathogenesis of ischemia-reperfusion injury (IRI) in a murine liver partial warm ischemia model, with emphasis on its regulation of hepatocyte death. RESULTS Rapamycin protected livers from IRI in the presence of fully developed liver inflammatory immune response. Rapamycin enhanced liver autophagy induction at the reperfusion stage. Dual mammalian (mechanistic) target of rapamycin (mTOR)1/2 inhibitor Torin 1, despite its ability to induced autophagy, failed to protect livers from IRI. The treatment with RPM, but not Torin 1, resulted in the enhanced activation of the mTORC2-Akt signaling pathway activation in livers after reperfusion. Inactivation of Akt by Triciribine abolished the liver protective effect of RPM. The differential cytoprotective effect of RPM and Torin 1 was confirmed in vitro in hepatocyte cultures. Rapamycin, but not Trin 1, protected hepatocytes from stress and tumor necrosis factor-α induced cell death; and inhibition of autophagy by chloroquine or Akt by Triciribine abolished RPM-mediated cytoprotection. CONCLUSION Rapamycin protected livers from IRI by both autophagy and mTORC2-Akt activation mechanisms.
Collapse
|
23
|
Kim H, Moon SY, Kim JS, Baek CH, Kim M, Min JY, Lee SK. Activation of AMP-activated protein kinase inhibits ER stress and renal fibrosis. Am J Physiol Renal Physiol 2015; 308:F226-36. [DOI: 10.1152/ajprenal.00495.2014] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It has been suggested that endoplasmic reticulum (ER) stress facilitates fibrotic remodeling. Therefore, modulation of ER stress may serve as one of the possible therapeutic approaches to renal fibrosis. We examined whether and how activation of AMP-activated protein kinase (AMPK) suppressed ER stress induced by chemical ER stress inducers [tunicamycin (TM) and thapsigargin (TG)] and also nonchemical inducers in tubular HK-2 cells. We further investigated the in vivo effects of AMPK on ER stress and renal fibrosis. Western blot analysis, immunofluorescence, small interfering (si)RNA experiments, and immunohistochemical staining were performed. Metformin (the best known clinical activator of AMPK) suppressed TM- or TG-induced ER stress, as shown by the inhibition of TM- or TG-induced upregulation of glucose-related protein (GRP)78 and phosphorylated eukaryotic initiation factor-2α through induction of heme oxygenase-1. Metformin inhibited TM- or TG-induced epithelial-mesenchymal transitions as well. Compound C (AMPK inhibitor) blocked the effect of metformin, and 5-aminoimidazole-4-carboxamide-1β riboside (another AMPK activator) exerted the same effects as metformin. Transfection with siRNA targeting AMPK blocked the effect of metformin. Consistent with the results of cell culture experiments, metformin reduced renal cortical GRP78 expression and increased heme oxygenase-1 expression in a mouse model of ER stress-induced acute kidney injury by TM. Activation of AMPK also suppressed ER stress by transforming growth factor-β, ANG II, aldosterone, and high glucose. Furthermore, metformin reduced GRP78 expression and renal fibrosis in a mouse model of unilateral ureteral obstruction. In conclusion, AMPK may serve as a promising therapeutic target through reducing ER stress and renal fibrosis.
Collapse
Affiliation(s)
- Hyosang Kim
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Soo Young Moon
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Joon-Seok Kim
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Chung Hee Baek
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Miyeon Kim
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Ji Yeon Min
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Sang Koo Lee
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| |
Collapse
|
24
|
Zhang C, Zheng L, Li L, Wang L, Li L, Huang S, Gu C, Zhang L, Yang C, Zhu T, Rong R. Rapamycin protects kidney against ischemia reperfusion injury through recruitment of NKT cells. J Transl Med 2014; 12:224. [PMID: 25134448 PMCID: PMC4145235 DOI: 10.1186/s12967-014-0224-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/04/2014] [Indexed: 11/10/2022] Open
Abstract
Background NKT cells play a protective role in ischemia reperfusion (IR) injury, of which the trafficking in the body and recruitment in injured organs can be influenced by immunosuppressive therapy. Therefore, we investigated the effects of rapamycin on kidneys exposed to IR injury in early stage and on trafficking of NKT cells in a murine model. Material and methods Balb/c mice were subjected to kidney 30 min ischemia followed by 24 h reperfusion. Rapamycin (2.5 ml/kg) was administered by gavage daily, starting 1 day before the operation. Renal function and histological changes were assessed. The proportion of NKT cells in peripheral blood, spleen and kidney was detected by flow cytometry. The chemokines and corresponding receptor involved in NKT cell trafficking were determined by RT-PCR and flow cytometry respectively. Results Rapamycin significantly improved renal function and ameliorated histological injury. In rapamycin-treated group, the proportion of NKT cells in spleen was significantly decreased but increased in peripheral blood and kidney. In addition, the CXCR3+ NKT cell in the kidney increased remarkably in the rapamycin-treated group. The chemokines, CXCL9 and CXCL10, as the ligands of CXCR3, were also increased in the rapamycin-treated kidney. Conclusions Rapamycin may recruit NKT cells from spleen to the IR-induced kidney to ameliorate renal IR injury in the early stage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | | | | |
Collapse
|
25
|
Wang L, Zhang Y, Yuan L, Liu C, Fu L, Mei C. Cyclin-dependent kinase inhibitor p18INK4c is involved in protective roles of heme oxygenase-1 in cisplatin-induced acute kidney injury. Int J Mol Med 2014; 34:911-7. [PMID: 24993528 DOI: 10.3892/ijmm.2014.1828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 06/26/2014] [Indexed: 11/05/2022] Open
Abstract
Experimental studies have demonstrated the protective effect of heme oxygenase (HO)-1 and cyclin‑dependent kinase inhibitors (CDKIs) in acute kidney injury (AKI), and it has been documented that some of the protective effect of HO-1 is mediated by CDKIs. However, the role of p18INK4c (p18), an inhibitor of CDK4 (INK4), which is a family member of CDKIs, has not been well characterized in kidney diseases. The aim of the present study was to demonstrate p18 protection from the relationship between p18 and HO-1 in cisplatin-induced AKI. Upregulation of p18 and HO-1 was demonstrated by quantitative polymerase chain reaction (qPCR) and western blotting in cisplatin-induced AKI in vitro and in vivo. The effect of HO-1 on p18 was determined by western blotting using the inducer and inhibitor of HO-1 in vitro. The potential effect of p18 on HO-1 in cisplatin‑induced AKI was examined by p18 gene knockout mice in vivo. The results showed that p18 and HO-1 were upregulated in cisplatin‑induced AKI in vitro and in vivo. Deletion of the p18 gene did not affect the basal and inducible expression of HO-1 in the AKI animals, while hemin (10 µM) and znpp (10 µM), the inducer and inhibitor, respectively, of HO-1, regulated p18 expression when incubated with the cells. The results indicated that p18 may play protective roles and may be associated with or partially account for the cytoprotective effects of HO-1 in cisplatin-induced AKI.
Collapse
Affiliation(s)
- Liang Wang
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, P.R. China
| | - Yi Zhang
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Li Yuan
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Chunyan Liu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116021, P.R. China
| | - Lili Fu
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| | - Changlin Mei
- Department of Nephrology, Changzheng Hospital, Shanghai 200003, P.R. China
| |
Collapse
|
26
|
|
27
|
Penicillinolide A: a new anti-inflammatory metabolite from the marine fungus Penicillium sp. SF-5292. Mar Drugs 2013; 11:4510-26. [PMID: 24225730 PMCID: PMC3853742 DOI: 10.3390/md11114510] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 10/30/2013] [Accepted: 10/31/2013] [Indexed: 01/04/2023] Open
Abstract
In the course of studies on bioactive metabolites from marine fungi, a new 10-membered lactone, named penicillinolide A (1) was isolated from the organic extract of Penicillium sp. SF-5292 as a potential anti-inflammatory compound. The structure of penicillinolide A (1) was mainly determined by analysis of NMR and MS data and Mosher’s method. Penicillinolide A (1) inhibited the production of NO and PGE2 due to inhibition of the expression of iNOS and COX-2. Penicillinolide A (1) also reduced TNF-α, IL-1β and IL-6 production, and these anti-inflammatory effects were shown to be correlated with the suppression of the phosphorylation and degradation of IκB-α, NF-κB nuclear translocation, and NF-κB DNA binding activity. In addition, using inhibitor tin protoporphyrin (SnPP), a competitive inhibitor of HO activity, it was verified that the inhibitory effects of compound 1 on the production of pro-inflammatory mediators and NF-κB DNA binding activity were partially associated with HO-1 expression through Nrf2 nuclear translocation.
Collapse
|
28
|
Kezic A, Thaiss F, Becker JU, Tsui TY, Bajcetic M. Effects of everolimus on oxidative stress in kidney model of ischemia/reperfusion injury. Am J Nephrol 2013; 37:291-301. [PMID: 23548777 DOI: 10.1159/000348496] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 01/29/2013] [Indexed: 01/30/2023]
Abstract
BACKGROUND/AIMS Reactive oxygen species play an important role in the pathogenesis of kidney ischemia/reperfusion injury (IRI) which may be influenced by immunosuppressive therapy. Pertinent to this, we investigated the effects of the mTOR inhibitor everolimus on redox settings and the activity of the anti-oxidative system in kidneys exposed to IRI. METHODS C57BL/6 mice were subjected to IRI by clamping both renal pedicles for 45 min. Everolimus was applied in daily, subcutaneous doses (0.25 mg/kg body weight), starting 1 day before IRI induction. Both everolimus-treated and non-treated mice were sacrificed at several time points, starting 30 min and finishing 7 days after IRI induction. Markers of oxidation such as glutathione and NADPH levels and anti-oxidative enzyme activities were determined in the kidneys. RESULTS In comparison to both sham and non-treated animals, the treatment with everolimus resulted in an increased level of markers of oxidation, including a lower level of glutathione, increased level of oxidized glutathione and reduced level of NADPH. The activity of superoxide dismutase was reduced in both experimental groups, but the effects were less pronounced in everolimus-treated animals. In the early phase of reperfusion, everolimus-treated animals showed higher activity of glutathione reductase in comparison to non-treated animals, whereas the activities of glutathione peroxidase and catalase were generally similar. The treatment with everolimus significantly reduced heme oxygenase-1 expression and increased iNOS mRNA expression when compared to non-treated animals. CONCLUSION Our data imply that everolimus treatment may decrease cytoprotective capacity in kidneys exposed to IRI due to promoted oxidative/nitrosative stress.
Collapse
|
29
|
Ptilovanciv EON, Fernandes GS, Teixeira LC, Reis LA, Pessoa EA, Convento MB, Simões MJ, Albertoni GA, Schor N, Borges FT. Heme oxygenase 1 improves glucoses metabolism and kidney histological alterations in diabetic rats. Diabetol Metab Syndr 2013; 5:3. [PMID: 23321053 PMCID: PMC3562196 DOI: 10.1186/1758-5996-5-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 01/09/2013] [Indexed: 01/08/2023] Open
Abstract
One important concern in the treatment of diabetes is the maintenance of glycemic levels and the prevention of diabetic nephropathy. Inducible heme oxygenase 1 (HO-1) is a rate-limiting enzyme thought to have antioxidant and cytoprotective roles. The goal of the present study was to analyze the effect of HO-1 induction in chronically hyperglycemic rats. The hyperglycemic rats were divided into two groups: one group, called STZ, was given a single injection of streptozotocin; and the other group was given a single streptozotocin injection as well as daily injections of hemin, an HO-1 inducer, over 60 days (STZ + HEME). A group of normoglycemic, untreated rats was used as the control (CTL).Body weight, diuresis, serum glucose levels, microalbuminuria, creatinine clearance rate, urea levels, sodium excretion, and lipid peroxidation were analyzed. Histological alterations and immunohistochemistry for HO-1 and inducible nitric oxide synthase (iNOS) were assessed. After 60 days, the STZ group exhibited an increase in blood glucose, diuresis, urea, microalbuminuria, and sodium excretion. There was no weight gain, and there was a decrease in creatinine clearance in comparison to the CTL group. In the STZ + HEME group there was an improvement in the metabolic parameters and kidney function, a decrease in blood glucose, serum urea, and microalbuminuria, and an increase of creatinine clearance, in comparison to the STZ group.There was glomerulosclerosis, collagen deposition in the STZ rats and increase in iNOS and HO-1 expression. In the STZ + HEME group, the glomerulosclerosis and fibrosis was prevented and there was an increase in the expression of HO-1, but decrease in iNOS expression and lipid peroxidation. In conclusion, our data suggest that chronic induction of HO-1 reduces hyperglycemia, improves glucose metabolism and, at least in part, protects the renal tissue from hyperglycemic injury, possibly through the antioxidant activity of HO-1.
Collapse
Affiliation(s)
- Ellen ON Ptilovanciv
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Gabryelle S Fernandes
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Luciana C Teixeira
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Luciana A Reis
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Edson A Pessoa
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Marcia B Convento
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Manuel J Simões
- Morphology Department, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Guilherme A Albertoni
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Nestor Schor
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Fernanda T Borges
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
30
|
Lee JH, Kim JH, Kim JS, Chang JW, Kim SB, Park JS, Lee SK. AMP-activated protein kinase inhibits TGF-β-, angiotensin II-, aldosterone-, high glucose-, and albumin-induced epithelial-mesenchymal transition. Am J Physiol Renal Physiol 2013; 304:F686-97. [PMID: 23324179 DOI: 10.1152/ajprenal.00148.2012] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a novel mechanism that promotes renal fibrosis. Transforming growth factor-β (TGF-β), angiotensin II, aldosterone, high glucose, and urinary albumin are well-known causes of EMT and renal fibrosis. We examined whether and how activation of AMP-activated protein kinase (AMPK) suppressed EMT induced by the above agents in tubular epithelial cells. All experiments were performed using HK-2 cells. Protein expression was measured by Western blot analysis. Intracellular reactive oxygen species (ROS) were analyzed by flow cytometry. Exposure of tubular cells to TGF-β (10 ng/ml), angiotensin II (1 μM), aldosterone (100 nM), high glucose (30 mM), and albumin (5 mg/ml) for 5 days induced EMT, as shown by upregulation of α-smooth muscle actin and downregulation of E-cadherin. ROS and NADPH oxidase 4 (Nox4) expression were increased, and antioxidants such as tiron and N-acetylcysteine inhibited EMT induction. Metformin (the best known clinical activator of AMPK) suppressed EMT induction through inhibition of ROS via induction of heme oxygenase-1 and endogenous antioxidant thioredoxin. An AMPK inhibitor (compound C) and AMPK small interfering RNA blocked the effect of metformin, and another AMPK activator [5-aminoimidazole-4-carboxamide-1β riboside (AICAR)] exerted the same effects as metformin. In conclusion, AMPK activation might be beneficial in attenuating the tubulointerstitial fibrosis induced by TGF-β, angiotensin II, aldosterone, high glucose, and urinary albumin.
Collapse
Affiliation(s)
- Jang Han Lee
- Department of Internal Medicine, Asan Institute for Life Sciences, College of Medicine, University of Ulsan, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Amano MT, Camara NOS. The immunomodulatory role of carbon monoxide during transplantation. Med Gas Res 2013; 3:1. [PMID: 23295066 PMCID: PMC3582539 DOI: 10.1186/2045-9912-3-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 12/31/2012] [Indexed: 01/03/2023] Open
Abstract
The number of organ and tissue transplants has increased worldwide in recent decades. However, graft rejection, infections due to the use of immunosuppressive drugs and a shortage of graft donors remain major concerns. Carbon monoxide (CO) had long been regarded solely as a poisonous gas. Ultimately, physiological studies unveiled the endogenous production of CO, particularly by the heme oxygenase (HO)-1 enzyme, recognizing CO as a beneficial gas when used at therapeutic doses. The protective properties of CO led researchers to develop uses for it, resulting in devices and molecules that can deliver CO in vitro and in vivo. The resulting interest in clinical investigations was immediate. Studies regarding the CO/HO-1 modulation of immune responses and their effects on various immune disorders gave rise to transplantation research, where CO was shown to be essential in the protection against organ rejection in animal models. This review provides a perspective of how CO modulates the immune system to improve transplantation and suggests its use as a therapy in the field.
Collapse
Affiliation(s)
- Mariane Tami Amano
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | |
Collapse
|
32
|
Nakagawa S, Nishihara K, Inui KI, Masuda S. Involvement of autophagy in the pharmacological effects of the mTOR inhibitor everolimus in acute kidney injury. Eur J Pharmacol 2012; 696:143-54. [DOI: 10.1016/j.ejphar.2012.09.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 09/05/2012] [Accepted: 09/17/2012] [Indexed: 10/27/2022]
|
33
|
Transcriptome analysis of renal ischemia/reperfusion injury and its modulation by ischemic pre-conditioning or hemin treatment. PLoS One 2012; 7:e49569. [PMID: 23166714 PMCID: PMC3498198 DOI: 10.1371/journal.pone.0049569] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 10/10/2012] [Indexed: 01/06/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) is a leading cause of acute renal failure. The definition of the molecular mechanisms involved in renal IRI and counter protection promoted by ischemic pre-conditioning (IPC) or Hemin treatment is an important milestone that needs to be accomplished in this research area. We examined, through an oligonucleotide microarray protocol, the renal differential transcriptome profiles of mice submitted to IRI, IPC and Hemin treatment. After identifying the profiles of differentially expressed genes observed for each comparison, we carried out functional enrichment analysis to reveal transcripts putatively involved in potential relevant biological processes and signaling pathways. The most relevant processes found in these comparisons were stress, apoptosis, cell differentiation, angiogenesis, focal adhesion, ECM-receptor interaction, ion transport, angiogenesis, mitosis and cell cycle, inflammatory response, olfactory transduction and regulation of actin cytoskeleton. In addition, the most important overrepresented pathways were MAPK, ErbB, JAK/STAT, Toll and Nod like receptors, Angiotensin II, Arachidonic acid metabolism, Wnt and coagulation cascade. Also, new insights were gained about the underlying protection mechanisms against renal IRI promoted by IPC and Hemin treatment. Venn diagram analysis allowed us to uncover common and exclusively differentially expressed genes between these two protective maneuvers, underscoring potential common and exclusive biological functions regulated in each case. In summary, IPC exclusively regulated the expression of genes belonging to stress, protein modification and apoptosis, highlighting the role of IPC in controlling exacerbated stress response. Treatment with the Hmox1 inducer Hemin, in turn, exclusively regulated the expression of genes associated with cell differentiation, metabolic pathways, cell cycle, mitosis, development, regulation of actin cytoskeleton and arachidonic acid metabolism, suggesting a pleiotropic effect for Hemin. These findings improve the biological understanding of how the kidney behaves after IRI. They also illustrate some possible underlying molecular mechanisms involved in kidney protection observed with IPC or Hemin treatment maneuvers.
Collapse
|
34
|
Elias RM, Correa-Costa M, Barreto CR, Silva RC, Hayashida CY, Castoldi Â, Gonçalves GM, Braga TT, Barboza R, Rios FJ, Keller AC, Cenedeze MA, Hyane MI, D'Império-Lima MR, Figueiredo-Neto AM, Reis MA, Marinho CRF, Pacheco-Silva A, Câmara NOS. Oxidative stress and modification of renal vascular permeability are associated with acute kidney injury during P. berghei ANKA infection. PLoS One 2012; 7:e44004. [PMID: 22952850 PMCID: PMC3432099 DOI: 10.1371/journal.pone.0044004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/27/2012] [Indexed: 12/21/2022] Open
Abstract
Malaria associated-acute kidney injury (AKI) is associated with 45% of mortality in adult patients hospitalized with severe form of the disease. However, the causes that lead to a framework of malaria-associated AKI are still poorly characterized. Some clinical studies speculate that oxidative stress products, a characteristic of Plasmodium infection, as well as proinflammatory response induced by the parasite are involved in its pathophysiology. Therefore, we aimed to investigate the development of malaria-associated AKI during infection by P. berghei ANKA, with special attention to the role played by the inflammatory response and the involvement of oxidative stress. For that, we took advantage of an experimental model of severe malaria that showed significant changes in the renal pathophysiology to investigate the role of malaria infection in the renal microvascular permeability and tissue injury. Therefore, BALB/c mice were infected with P. berghei ANKA. To assess renal function, creatinine, blood urea nitrogen, and ratio of proteinuria and creatininuria were evaluated. The products of oxidative stress, as well as cytokine profile were quantified in plasma and renal tissue. The change of renal microvascular permeability, tissue hypoxia and cellular apoptosis were also evaluated. Parasite infection resulted in renal dysfunction. Furthermore, we observed increased expression of adhesion molecule, proinflammatory cytokines and products of oxidative stress, associated with a decrease mRNA expression of HO-1 in kidney tissue of infected mice. The measurement of lipoprotein oxidizability also showed a significant increase in plasma of infected animals. Together, our findings support the idea that products of oxidative stress, as well as the immune response against the parasite are crucial to changes in kidney architecture and microvascular endothelial permeability of BALB/c mice infected with P. berghei ANKA.
Collapse
Affiliation(s)
- Rosa Maria Elias
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Matheus Correa-Costa
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Reinaldo Correia Silva
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caroline Y. Hayashida
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Ângela Castoldi
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Giselle Martins Gonçalves
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Tarcio Teodoro Braga
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Renato Barboza
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Francisco José Rios
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Marcos Antonio Cenedeze
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Meire Ioshie Hyane
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Maria Regina D'Império-Lima
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Marlene Antônia Reis
- Divisão de Patologia, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | | | - Alvaro Pacheco-Silva
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto Israelita de Ensino e Pesquisa Albert Einstein, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
35
|
Correa-Costa M, Amano MT, Câmara NOS. Cytoprotection behind heme oxygenase-1 in renal diseases. World J Nephrol 2012; 1:4-11. [PMID: 24175236 PMCID: PMC3782207 DOI: 10.5527/wjn.v1.i1.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 10/27/2011] [Accepted: 12/27/2011] [Indexed: 02/06/2023] Open
Abstract
Renal insults are considered a public health problem and are linked to increased rates of morbidity and mortality worldwide. The heme oxygenase (HO) system consists of evolutionary specialized machinery that degrades free heme and produces carbon monoxide, biliverdin and free iron. In this sense, the inducible isoform HO-1 seems to develop an important role and is widely studied. The reaction involved with the HO-1 molecule provides protection to injured tissue, directly by reducing the toxic heme molecule and indirectly by the release of its byproducts. The up regulation of HO-1 enzyme has largely been described as providing antioxidant, antiapoptotic, anti-inflammatory and immunomodulatory properties. Several works have explored the importance of HO-1 in renal diseases and they have provided consistent evidence that its overexpression has beneficial effects in such injuries. So, in this review we will focus on the role of HO-1 in kidney insults, exploring the protective effects of its up regulation and the enhanced deleterious effects of its inhibition or gene deletion.
Collapse
Affiliation(s)
- Matheus Correa-Costa
- Matheus Correa-Costa, Mariane Tami Amano, Niels Olsen Saraiva Câmara, Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, 05508-000, São Paulo, Brazil
| | | | | |
Collapse
|
36
|
Hara Y, Stolk M, Ringe J, Dehne T, Ladhoff J, Kotsch K, Reutzel-Selke A, Reinke P, Volk HD, Seifert M. In vivo effect of bone marrow-derived mesenchymal stem cells in a rat kidney transplantation model with prolonged cold ischemia. Transpl Int 2011; 24:1112-23. [PMID: 21880071 DOI: 10.1111/j.1432-2277.2011.01328.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Brain death and prolonged cold ischemia are major contributors to the poorer long-term outcome of transplants from deceased donor kidney transplants, with an even higher impact if expanded criteria donors ('marginal organs') are used. Targeting ischemia-reperfusion injury-related intragraft inflammation is an attractive concept to improve the outcome of those grafts. As mesenchymal stem cells (MSCs) express both immunomodulatory and tissue repair properties, we evaluated their therapeutic efficacy in a rat kidney transplant model of prolonged cold ischemia. The in vitro immunomodulatory capacity of bone marrow-derived rat MSCs was tested in co-cultures with rat lymph node cells. For in vivo studies, Dark Agouti rat kidneys were cold preserved and transplanted into Lewis rats. Syngeneic Lewis MSCs were administered intravenously. Transplants were harvested on day 3, and inflammation was examined by quantitative RT-PCR and histology. Similarly to MSCs from other species, rat MSCs in vitro also showed a dose-dependent immunomodulatory capacity. Most importantly, in vivo administration of MSCs reduced the intragraft gene expression of different pro-inflammatory cytokines, chemokines, and intercellular adhesion molecule-1. In addition, fewer antigen-presenting cells were recruited into the renal allograft. In conclusion, rat MSCs ameliorate inflammation induced by prolonged cold ischemia in kidney transplantation.
Collapse
Affiliation(s)
- Yoshiaki Hara
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cicora F, Lausada N, Vasquez DN, Cicora P, Guerrieri D, Gonzalez P, Zalazar G, Stringa P, Raimondi C. Protective effect of immunosuppressive treatment before orthotopic kidney autotransplantation. Transpl Immunol 2011; 24:107-12. [DOI: 10.1016/j.trim.2010.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 10/12/2010] [Accepted: 10/19/2010] [Indexed: 12/27/2022]
|
38
|
Correa-Costa M, Semedo P, Monteiro APFS, Silva RC, Pereira RL, Gonçalves GM, Marques GDM, Cenedeze MA, Faleiros ACG, Keller AC, Shimizu MHM, Seguro AC, Reis MA, Pacheco-Silva A, Câmara NOS. Induction of heme oxygenase-1 can halt and even reverse renal tubule-interstitial fibrosis. PLoS One 2010; 5:e14298. [PMID: 21179206 PMCID: PMC3001459 DOI: 10.1371/journal.pone.0014298] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Accepted: 11/23/2010] [Indexed: 01/08/2023] Open
Abstract
Background The tubule-interstitial fibrosis is the hallmark of progressive renal disease and is strongly associated with inflammation of this compartment. Heme-oxygenase-1 (HO-1) is a cytoprotective molecule that has been shown to be beneficial in various models of renal injury. However, the role of HO-1 in reversing an established renal scar has not yet been addressed. Aim We explored the ability of HO-1 to halt and reverse the establishment of fibrosis in an experimental model of chronic renal disease. Methods Sprague-Dawley male rats were subjected to unilateral ureteral obstruction (UUO) and divided into two groups: non-treated and Hemin-treated. To study the prevention of fibrosis, animals were pre-treated with Hemin at days -2 and -1 prior to UUO. To investigate whether HO-1 could reverse established fibrosis, Hemin therapy was given at days 6 and 7 post-surgery. After 7 and/or 14 days, animals were sacrificed and blood, urine and kidney tissue samples were collected for analyses. Renal function was determined by assessing the serum creatinine, inulin clearance, proteinuria/creatininuria ratio and extent of albuminuria. Arterial blood pressure was measured and fibrosis was quantified by Picrosirius staining. Gene and protein expression of pro-inflammatory and pro-fibrotic molecules, as well as HO-1 were performed. Results Pre-treatment with Hemin upregulated HO-1 expression and significantly reduced proteinuria, albuminuria, inflammation and pro-fibrotic protein and gene expressions in animals subjected to UUO. Interestingly, the delayed treatment with Hemin was also able to reduce renal dysfunction and to decrease the expression of pro-inflammatory molecules, all in association with significantly reduced levels of fibrosis-related molecules and collagen deposition. Finally, TGF-β protein production was significantly lower in Hemin-treated animals. Conclusion Treatment with Hemin was able both to prevent the progression of fibrosis and to reverse an established renal scar. Modulation of inflammation appears to be the major mechanism behind HO-1 cytoprotection.
Collapse
Affiliation(s)
- Matheus Correa-Costa
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo, Brazil
| | - Patricia Semedo
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ana Paula F. S. Monteiro
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Reinaldo C. Silva
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Rafael L. Pereira
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Giselle M. Gonçalves
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo, Brazil
| | - Georgia Daniela Marcusso Marques
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Marcos A. Cenedeze
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ana C. G. Faleiros
- Pathology Division, Federal University of Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Alexandre C. Keller
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Maria H. M. Shimizu
- Nephrology Department, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Antônio C. Seguro
- Nephrology Department, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Marlene A. Reis
- Pathology Division, Federal University of Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Alvaro Pacheco-Silva
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Niels O. S. Câmara
- Laboratory of Clinical and Experimental Immunology, Nephrology Division, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo, Brazil
- * E-mail:
| |
Collapse
|
39
|
Martinez-Palli G, Hirose R, Liu T, Xu F, Dang K, Feiner J, Serkova NJ, Niemann CU. Donor pre-treatment with everolimus or cyclosporine does not reduce ischaemia-reperfusion injury in a rat kidney transplant model. Nephrol Dial Transplant 2010; 26:1813-20. [PMID: 21068143 DOI: 10.1093/ndt/gfq646] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Immunosuppressive agents have been investigated in renal ischaemia-reperfusion injury (IRI) and have frequently demonstrated a beneficial effect. Most studies focused on treatment of the recipient at the time of transplantation. Pre-treatment of these organs before injury (pharmacological pre-conditioning) may particularly protect these organs. This study aimed to investigate the possible protective effects of donor pre-treatment with cyclosporine (CsA) or the mTOR inhibitor everolimus or their combination against IRI during renal transplantation in a rat model. METHODS Donors received vehicle, CsA (5 mg/kg), everolimus (0.5 mg/kg) or CsA + everolimus. Two oral doses were administered to the donors at 24 h and again at 6 h prior to donor kidney removal. Syngeneic rat kidneys were preserved in UW solution for 24 h prior to transplantation. After 24 h of reperfusion, blood and tissue samples were collected from recipients for further analysis. RESULTS Renal functions as determined by creatinine and necrosis scores were not different between the experimental groups. Cleaved caspase-3, heat shock protein 70 (HSP70), tumor-necrosis factor-alpha (TNF-α) and nitrotyrosine protein levels were not statistically different between the four treatment groups at 24 h post-transplantation. Blood NMR analysis on metabolic markers for IRI reveals no beneficial effects of donor pre-treatment on the 24-h outcome in transplantation. CONCLUSIONS When given alone or as a combination to donors before organ recovery, cyclosporine or everolimus does not appear to ameliorate IRI.
Collapse
Affiliation(s)
- Graciela Martinez-Palli
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Sonoda H, Prachasilchai W, Kondo H, Yokota-Ikeda N, Oshikawa S, Ito K, Ikeda M. The protective effect of radicicol against renal ischemia--reperfusion injury in mice. J Pharmacol Sci 2010; 112:242-6. [PMID: 20093793 DOI: 10.1254/jphs.09259sc] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Overexpression of heat shock protein 70 kDa (HSP70) is known to confer cellular protection against ischemia-reperfusion (I/R) injury. Radicicol, a HSP90 inhibitor, has been reported to induce the expression of HSP70 protein. Here we studied whether radicicol attenuated renal I/R injury in vivo. Treatment of mice with radicicol ameliorated renal I/R injury and increased renal HSP70 mRNA and protein. Administration of radicicol with quercetin, an inhibitor of HSP70 induction, eliminated the renoprotective effect of radicicol. Our results suggest that the up-regulation of renal HSP70 protein by radicicol leads to a novel drug therapy against renal I/R injury.
Collapse
Affiliation(s)
- Hiroko Sonoda
- Department of Veterinary Pharmacology, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Säemann MD, Haidinger M, Hecking M, Hörl WH, Weichhart T. The multifunctional role of mTOR in innate immunity: implications for transplant immunity. Am J Transplant 2009; 9:2655-61. [PMID: 19788500 DOI: 10.1111/j.1600-6143.2009.02832.x] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The mammalian target of rapamycin (mTOR) is an evolutionary conserved serine-threonine kinase that senses various environmental stimuli in most cells primarily to control cell growth. Restriction of cellular proliferation by mTOR inhibition led to the use of mTOR inhibitors as immunosuppressants in allogeneic transplantation as well as novel anticancer agents. However, distinct inflammatory side effects such as fever, pneumonitis, glomerulonephritis or anemia of chronic disease have been observed under this treatment regime. Apart from the mere cell-cycle regulatory effect of mTOR in dividing cells, recent data revealed a master regulatory role of mTOR in the innate immune system. Hence, inhibition of mTOR promotes proinflammatory cytokines such as IL-12 and IL-1beta, inhibits the anti-inflammatory cytokine IL-10 and boosts MHC antigen presentation via autophagy in monocytes/macrophages and dendritic cells. Moreover, mTOR regulates type I interferon production and the expression of chemokine receptors and costimulatory molecules. These results place mTOR in a complex immunoregulatory context by controlling innate and adaptive immune responses. In this review, we discuss the clinical consequences of mTOR-inhibitor therapy and aim to integrate this recent data into our current view of the molecular mechanisms of clinically employed mTOR inhibitors and discuss their relevance with special emphasis to transplantation.
Collapse
Affiliation(s)
- M D Säemann
- Clinical Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria.
| | | | | | | | | |
Collapse
|
42
|
The anti-inflammatory effect of tussilagone, from Tussilago farfara, is mediated by the induction of heme oxygenase-1 in murine macrophages. Int Immunopharmacol 2009; 9:1578-84. [DOI: 10.1016/j.intimp.2009.09.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 09/12/2009] [Accepted: 09/23/2009] [Indexed: 01/26/2023]
|
43
|
Abraham NG, Cao J, Sacerdoti D, Li X, Drummond G. Heme oxygenase: the key to renal function regulation. Am J Physiol Renal Physiol 2009; 297:F1137-52. [PMID: 19570878 PMCID: PMC2781329 DOI: 10.1152/ajprenal.90449.2008] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 06/09/2009] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase (HO) plays a critical role in attenuating the production of reactive oxygen species through its ability to degrade heme in an enzymatic process that leads to the production of equimolar amounts of carbon monoxide and biliverdin/bilirubin and the release of free iron. The present review examines the beneficial role of HO-1 (inducible form of HO) that is achieved by increased expression of this enzyme in renal tissue. The influence of the HO system on renal physiology, obesity, vascular dysfunction, and blood pressure regulation is reviewed, and the clinical potential of increased levels of HO-1 protein, HO activity, and HO-derived end products of heme degradation is discussed relative to renal disease. The use of pharmacological and genetic approaches to investigate the role of the HO system in the kidney is key to the development of therapeutic approaches to prevent the adverse effects that accrue due to an impairment in renal function.
Collapse
Affiliation(s)
- Nader G Abraham
- New York Medical College, Department of Pharmacology, Valhalla, NY 10595, USA.
| | | | | | | | | |
Collapse
|
44
|
Suyani E, Derici UB, Sahin T, Ofluoglu E, Pasaoglu H, Erdem O, Barit G, Reis KA, Erten Y, Arinsoy T, Sindel S. Effects of Everolimus on Cytokines, Oxidative Stress, and Renal Histology in Ischemia-Reperfusion Injury of the Kidney. Ren Fail 2009; 31:698-703. [PMID: 19814637 DOI: 10.3109/08860220903134555] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Elif Suyani
- Gazi University, Department of Internal Medicine, 06510, Besevler, Ankara, Turkey.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Feitoza CQ, Semedo P, Gonçalves GM, Cenedeze MA, Pinheiro HS, Dos Santos OFP, Landgraf RG, Pacheco-Silva A, Câmara NOS. Modulation of inflammatory response by selective inhibition of cyclooxygenase-1 and cyclooxygenase-2 in acute kidney injury. Inflamm Res 2009; 59:167-75. [PMID: 19711010 DOI: 10.1007/s00011-009-0083-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 08/07/2009] [Accepted: 08/10/2009] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE AND DESIGN This work explored the role of inhibition of cyclooxygenases (COXs) in modulating the inflammatory response triggered by acute kidney injury. MATERIAL C57Bl/6 mice were used. TREATMENT Animals were treated or not with indomethacin (IMT) prior to injury (days -1 and 0). METHODS Animals were subjected to 45 min of renal pedicle occlusion and sacrificed at 24 h after reperfusion. Serum creatinine and blood urea nitrogen, reactive oxygen species (ROS), kidney myeloperoxidase (MPO) activity, and prostaglandin E2 (PGE(2)) levels were analyzed. Tumor necrosis factor (TNF)-alpha, t-bet, interleukin (IL)-10, IL-1beta, heme oxygenase (HO)-1, and prostaglandin E synthase (PGES) messenger RNA (mRNA) were studied. Cytokines were quantified in serum. RESULTS IMT-treated animals presented better renal function with less acute tubular necrosis and reduced ROS and MPO production. Moreover, the treatment was associated with lower expression of TNF-alpha, PGE(2), PGES, and t-bet and upregulation of HO-1 and IL-10. This profile was mirrored in serum, where inhibition of COXs significantly decreased interferon (IFN)-gamma, TNF-alpha, and IL-12 p70 and upregulated IL-10. CONCLUSIONS COXs seem to play an important role in renal ischemia and reperfusion injury, involving the secretion of pro-inflammatory cytokines, activation of neutrophils, and ROS production. Inhibition of COX pathway is intrinsically involved with cytoprotection.
Collapse
Affiliation(s)
- Carla Q Feitoza
- Laboratory of Experimental and Clinical Immunology, Nephrology Division, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Therapeutic role of sirolimus in non-transplant kidney disease. Pharmacol Ther 2009; 123:187-206. [DOI: 10.1016/j.pharmthera.2009.03.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Accepted: 03/24/2009] [Indexed: 12/20/2022]
|
47
|
Hassan IR, Gronert K. Acute changes in dietary omega-3 and omega-6 polyunsaturated fatty acids have a pronounced impact on survival following ischemic renal injury and formation of renoprotective docosahexaenoic acid-derived protectin D1. THE JOURNAL OF IMMUNOLOGY 2009; 182:3223-32. [PMID: 19234220 DOI: 10.4049/jimmunol.0802064] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Exacerbated inflammation plays an important role in the pathogenesis of ischemic renal injury (IRI), which is the major cause of intrinsic acute renal failure. Clinical studies suggest that long-term treatment with omega-3 polyunsaturated fatty acids (PUFA) improves renal function and lowers the risk of death or end-stage renal disease. Docosahexaenoic acid, a principle omega-3 PUFA of fish oils, is of particular interest as it is found in most human tissues and is converted to protectin D1 (PD1), which exhibits antiinflammatory and proresolving bioactions. We set out to investigate the impact of acute dietary modulation of omega-3 or omega-6 PUFA on IRI and renal lipid autacoid circuits, using an established mouse model and liquid chromatography-mass spectroscopy/mass spectroscopy-based lipidomics. Thirty minutes of renal ischemia significantly elevated serum creatinine in the omega-6 diet group while renal function remained normal in the matched omega-3 diet group. Notably, extending ischemia to 45 min caused 100% mortality in the omega-6 group, in sharp contrast to 0% mortality in the omega-3 group. Protection against IRI in the omega-3 group correlated with decreased polymorphonuclear leukocyte recruitment, chemokine and cytokine levels, abrogated formation of lipoxygenase- and cyclooxygenase-derived eicosanoids, and increased renal levels of PD1. Systemic treatment with PD1 reduced kidney polymorphonuclear leukocyte influx and, more importantly, amplified renoprotective heme-oxygenase-1 protein and mRNA expression in injured and uninjured kidneys. These findings suggest therapeutic or dietary amplification of PD1 circuits restrains acute renal injury and that short-term changes in dietary omega-3 and omega-6 PUFA dramatically impacts renal lipid autacoid formation and outcome of IRI.
Collapse
Affiliation(s)
- Iram R Hassan
- New York Medical College, Department of Pharmacology, Valhalla, NY 10595, USA
| | | |
Collapse
|
48
|
Jiang K, Cheng L, Wang J, Li J, Nie J. Heme oxygenase-1 expression in rats with acute lung rejection and implication. ACTA ACUST UNITED AC 2009; 29:84-7. [PMID: 19224170 DOI: 10.1007/s11596-009-0118-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Indexed: 01/03/2023]
Abstract
This study investigated the expression of hemeoxygenase-1 (HO-1) in rats with acute lung rejection and its implication. A valid rat orthotopic left lung transplantation model (SD rat-->Wistar rat) was established by using an improved three-cuff anastomosis technique. The rats were divided into control group, CoPP (HO-1 inducer)-treated group and ZnPP (HO-1 inhibitor)- treated group. The severity of acute rejection was graded on the basis of the morphologic changes of the lung samples stained with HE. The expression of HO-1 protein in lung tissue was detected by using immunohistochemistry and Western blot, and HO-1 mRNA activity was assayed by RT-PCR. The results showed that the expression of HO-1 protein was significantly increased with the acute rejection grading in rats (P<0.01). As compared with control and ZnPP-treated groups, the severity of acute rejection was not alleviated and the grade not reduced significantly in CoPP-treated group (P>0.05). It was concluded that HO-1 protein might be involved in the pathological process of post-graft acute rejection. The expression of HO-1 protein was increased gradually with aggravation of acute rejection, and HO-1 protein might be used as an index to monitor acute rejection after lung transplantation.
Collapse
Affiliation(s)
- Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | |
Collapse
|
49
|
Cudratricusxanthone A from Cudrania tricuspidata suppresses pro-inflammatory mediators through expression of anti-inflammatory heme oxygenase-1 in RAW264.7 macrophages. Int Immunopharmacol 2009; 9:241-6. [DOI: 10.1016/j.intimp.2008.11.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 11/14/2008] [Accepted: 11/20/2008] [Indexed: 12/17/2022]
|
50
|
Cao J, Inoue K, Li X, Drummond G, Abraham NG. Physiological significance of heme oxygenase in hypertension. Int J Biochem Cell Biol 2008; 41:1025-33. [PMID: 19027871 DOI: 10.1016/j.biocel.2008.10.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 10/21/2008] [Accepted: 10/27/2008] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed an explosion in the elucidation of the role that the heme oxygenase system plays in human physiology. This system encompasses not only the heme degradative pathway, including heme oxygenase and biliverdin reductase, but also the products of heme degradation, carbon monoxide, iron, and biliverdin/bilirubin. Their role in diabetes, inflammation, heart disease, hypertension, transplantation, and pulmonary disease are areas of burgeoning research. The research has focused not only on heme itself but also on its metabolic products as well as endogenous compounds involved in a vast number of genetic and metabolic processes that are affected when heme metabolism is perturbed. It should be noted, however, that although the use of carbon monoxide and biliverdin/bilirubin as therapeutic agents has been successful, these agents can be toxic at high levels in tissue, e.g., kernicterus. Care must be used to ensure that when these compounds are used as therapeutic agents their deleterious effects are minimized or avoided. On balance, however, the strategies to target heme oxygenase-1 as described in this review offer promising therapeutic approaches to clinicians for the effective management of hypertension and renal function. The approaches detailed may prove to be seminal in the development of a new therapeutic strategy to treat hypertension.
Collapse
Affiliation(s)
- Jian Cao
- New York Medical College, Department of Pharmacology, Valhalla, NY 10595, United States
| | | | | | | | | |
Collapse
|