1
|
Sun W, Choi HS, Kim CS, Bae EH, Ma SK, Kim SW. Maslinic Acid Attenuates Ischemia/Reperfusion-Induced Acute Kidney Injury by Suppressing Inflammation and Apoptosis Through Inhibiting NF-κB and MAPK Signaling Pathway. Front Pharmacol 2022; 13:807452. [PMID: 35496304 PMCID: PMC9039024 DOI: 10.3389/fphar.2022.807452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/03/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation and apoptosis are the major contributors to the mechanisms of acute kidney injury (AKI) due to renal ischemia-reperfusion injury (IRI). Maslinic acid (MA), a pentacyclic triterpene acid mostly found in dietary plants, the current study was to demonstrate the renoprotective effect of MA on IRI-induced AKI, and to investigate the role of inflammation and apoptosis-related signaling pathways as a molecular mechanism. C57BL/6J mice were subjected to IRI for 72 h, and MA was daily administered by intraperitoneal injection during this period. In parallel, rat renal proximal tubule cells (NRK52E) were prophylactically treated with MA and then exposed to hydrogen peroxide (H2O2). MA treatment significantly inhibited the mRNA expression of interleukin (IL-1β), tumor necrosis factor-α (TGF-α), monocyte chemoattractant protein-1 (MCP-1), and intercellular adhesion molecule-1(ICAM-1). Also, MA reduced the expression of Bax/Bcl2 ratio and cleaved caspase-3. In NRK52 cells, MA inhibited the IκBα degradation, blocked NF-κB/p65 phosphorylation, and nuclear translocation. The phosphorylation of ERK, JNK, and p38 was attenuated by MA in IRI-induced kidney injury and H2O2-stimulated NRK52 cells. The expression levels of IL-1β, MCP-1, and ICAM-1 were upregulated in H2O2-stimulated NRK52E cells, which was attenuated by NF-κB inhibitor. H2O2 treatment increased the Bax/Bcl2 ratio and cleaved caspase-3 in NRK52E cells, which was counteracted by MAPK inhibitors. Together, our data demonstrate that MA suppresses IR-induced AKI injury through NF-κB and MAPK signaling pathways and that MA is a promising agent in the treatment of kidney diseases.
Collapse
|
2
|
Zhou S, Guo J, Liao X, Zhou Q, Qiu X, Jiang S, Xu N, Wang X, Zhao L, Hu W, Xie L, Xie P, Cui Y, Yang Y, Patzak A, Persson PB, Mao J, Lai EY. rhADAMTS13 reduces oxidative stress by cleaving VWF in ischaemia/reperfusion-induced acute kidney injury. Acta Physiol (Oxf) 2022; 234:e13778. [PMID: 34989474 DOI: 10.1111/apha.13778] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/17/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022]
Abstract
AIMS Acute kidney injury (AKI), a major health burden, lacks effective therapy. Anti-inflammatory actions of a disintegrin and metalloproteinase with a thrombospondin type 1 motif member 13 (ADAMTS13) may provide a new treatment option for AKI. Along with inflammation, oxidative stress is critical for AKI development, yet the impact of ADAMTS13 on oxidative stress in AKI remains to be fully elucidated. METHODS We assess recombinant human ADAMTS13 (rhADAMTS13) actions on oxidative stress in a murine ischaemia/reperfusion (IR) model. Antioxidant stress-enzyme activities, renal morphology, kidney function markers and vascular function of isolated afferent arterioles are quantified. RESULTS rhADAMTS13 provided after IR, reduces blood urea nitrogen (BUN) by 33% and serum creatinine (Scr) by 73% in 24 hours post-IR. rhADAMTS13 reduces BUN (40.03 ± 20.34 mmol/L vs 72.35 ± 18.74 mmol/L, P < .01), Scr (75.67 ± 51.19 μmol/L vs 176.17 ± 55.38 μmol/L, P < .01) and proteinuria by 41% in 48 hours post-IR as well. Moreover, rhADAMTS13 administration decreases malondialdehyde (MDA) and increases the activity of antioxidant stress enzymes, and attenuates reactive oxygen species production. rhADAMTS13 also upregulates nuclear factor-erythroid-2-related factor 2/haem oxygenase-1, enhances antioxidant enzymes activity and alleviates endothelial dysfunction. Finally, treatment with rhADAMTS13 mitigates severe functional and morphological injury present in IR mice. Extracellular signal-regulated kinase (ERK) phosphorylation is limited by rhADAMTS13 and PPARγ expression is partly restored in ischaemic kidneys. Co-administration of von Willebrand factor (VWF) impairs rhADAMTS13's antioxidant capacity and its protective role in IR. CONCLUSION rhADAMTS13 alleviates renal IR injury through antioxidant effects by cleaving VWF.
Collapse
Affiliation(s)
- Suhan Zhou
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Jie Guo
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Xinxin Liao
- Department of Anesthesiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Qin Zhou
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Xingyu Qiu
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Nephrology Center of Kidney and Urology the Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen China
| | - Nan Xu
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
- Department of Pathophysiology School of Basic Medical Sciences Henan University Kaifeng China
| | - Xiaohua Wang
- Department of Nephrology Center of Kidney and Urology the Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen China
| | - Liang Zhao
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Weipeng Hu
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Lanyu Xie
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Peng Xie
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Yu Cui
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Yi Yang
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Andreas Patzak
- Charité–Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Pontus B. Persson
- Charité–Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Jianhua Mao
- Department of Nephrology the Children's Hospital of Zhejiang University School of Medicine Hangzhou China
| | - En Yin Lai
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
- Department of Nephrology Center of Kidney and Urology the Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen China
- Charité–Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
3
|
Ou YC, Li JR, Wu CC, Yu TM, Chen WY, Liao SL, Kuan YH, Chen YF, Chen CJ. Cadmium induces the expression of Interleukin-6 through Heme Oxygenase-1 in HK-2 cells and Sprague-Dawley rats. Food Chem Toxicol 2022; 161:112846. [PMID: 35122928 DOI: 10.1016/j.fct.2022.112846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/06/2022] [Accepted: 01/29/2022] [Indexed: 11/15/2022]
Abstract
Cadmium is toxic to the kidney through mechanisms involving oxidative stress and inflammation. We studied reciprocal crosstalk among the oxidative stress, inflammation, and the nuclear Nrf2 pathway in cadmium-induced nephrotoxicity on HK-2 human renal proximal tubular epithelial cells. Cadmium chloride (CdCl2) caused cell viability loss, Reactive Oxygen Species (ROS) generation, glutathione reduction, and Interleukin-6 (IL-6) expression, accompanied by Nrf2 activation and Heme Oxygenase-1 (HO-1) expression. Pharmacological treatments demonstrated cytotprotective and anti-inflammatory effects of Nrf2 activation. Intriguingly, inhibition of HO-1 activity mitigated cell viability loss and IL-6 expression in CdCl2-treated cells. Parallel attenuation by HO-1 inhibitor was demonstrated in cadmium-induced ROS generation and glutathione reduction. CdCl2-treated cells also increased levels of ferrous iron, cGMP, Mitogen-Activated Protein Kinases phosphorylation, as well as NF-κB DNA-binding activity. These increments were mitigated by antioxidant N-Acetyl Cysteine, HO-1 inhibitor SnPP, and PKG inhibitor KT5823, and were mimicked by the Carbon Monoxide-releasing compound. In the kidney cortex of CdCl2-exposed Sprague-Dawley rats, we found similar renal injury, histological changes, ROS generation, IL-6 expression, and accompanied pro-oxidant and pro-inflammatory changes. These observations indicated that cadmium-induced nephrotoxicity was associated with oxidative stress and inflammation, and HO-1 likely acts as a linking molecule to induce nephrotoxicity-associated IL-6 expression upon cadmium exposure.
Collapse
Affiliation(s)
- Yen-Chuan Ou
- Department of Urology, Tungs' Taichung MetroHarbor Hospital, Taichung City, Taiwan
| | - Jian-Ri Li
- Division of Urology, Taichung City, Taiwan; Department of Nursing, HungKuang University, Taichung City, Taiwan
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung City, Taiwan; Department of Financial Engineering, Providence University, Taichung City, Taiwan; Department of Data Science and Big Data Analytics, Providence University, Taichung City, Taiwan
| | - Tung-Min Yu
- Division of Nephrology, Taichung City, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung City, Taiwan
| | - Yu-Fan Chen
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City, Taiwan.
| |
Collapse
|
4
|
Hu W, Jiang S, Liao Y, Li J, Dong F, Guo J, Wang X, Fei L, Cui Y, Ren X, Xu N, Zhao L, Chen L, Zheng Y, Li L, Patzak A, Persson PB, Zheng Z, Lai EY. High phosphate impairs arterial endothelial function through AMPK-related pathways in mouse resistance arteries. Acta Physiol (Oxf) 2021; 231:e13595. [PMID: 33835704 DOI: 10.1111/apha.13595] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/10/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
AIMS In patients with renal disease, high serum phosphate shows a relationship with cardiovascular risk. We speculate that high phosphate (HP) impairs arterial vasodilation via the endothelium and explore potential underlying mechanisms. METHODS Isolated vessel relaxation, endothelial function, glomerular filtration rate (GFR), oxidative stress status and protein expression were assessed in HP diet mice. Mitochondrial function and protein expression were assessed in HP-treated human umbilical vein endothelial cells (HUVECs). RESULTS High phosphate (1.3%) diet for 12 weeks impaired endothelium-dependent relaxation in mesenteric arteries, kidney interlobar arteries and afferent arterioles; reduced GFR and the blood pressure responses to acute administration of acetylcholine. The PPARα/LKB1/AMPK/eNOS pathway was attenuated in the endothelium of mesenteric arteries from HP diet mice. The observed vasodilatory impairment of mesenteric arteries was ameliorated by PPARα agonist WY-14643. The phosphate transporter PiT-1 knockdown prevented HP-mediated suppression of eNOS activity by impeding phosphorus influx in HUVECs. Endothelium cytoplasmic and mitochondrial reactive oxygen species (ROS) were increased in HP diet mice. Moreover HP decreased the expression of mitochondrial-related antioxidant genes. Finally, mitochondrial membrane potential and PGC-1α expression were reduced by HP treatment in HUVECs, which was partly restored by AMPKα agonist. CONCLUSIONS HP impairs endothelial function by reducing NO bioavailability via decreasing eNOS activity and increasing mitochondrial ROS, in which the AMPK-related signalling pathways may play a key role.
Collapse
Affiliation(s)
- Weipeng Hu
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Yixin Liao
- Department of Obstetrics and Gynecology Nanfang HospitalSouthern Medical University Guangzhou China
| | - Jinhong Li
- Department of Nephrology Center of Kidney The Seventh Affiliate HospitalSun Yat‐sen University Shenzhen China
| | - Fang Dong
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Jie Guo
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Xiaohua Wang
- Department of Nephrology Center of Kidney The Seventh Affiliate HospitalSun Yat‐sen University Shenzhen China
| | - Lingyan Fei
- Department of Nephrology Center of Kidney The Seventh Affiliate HospitalSun Yat‐sen University Shenzhen China
| | - Yu Cui
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Xiaoqiu Ren
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Nan Xu
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Liang Zhao
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
- Department of Physiology School of Basic Medical Sciences Guangzhou Medical University Guangzhou China
| | - Limeng Chen
- Department of Nephrology Peking Union Medical College HospitalChinese Academy of Medical Science & Peking Union Medical College Beijing China
| | - Yali Zheng
- Department of Nephrology Ningxia people’s hospital Yinchuan China
| | - Lingli Li
- Division of Nephrology and Hypertension Georgetown University Washington DC USA
| | - Andreas Patzak
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Pontus B. Persson
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Zhihua Zheng
- Department of Nephrology Center of Kidney The Seventh Affiliate HospitalSun Yat‐sen University Shenzhen China
| | - En Yin Lai
- Department of Physiology School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
- Department of Nephrology Center of Kidney The Seventh Affiliate HospitalSun Yat‐sen University Shenzhen China
- Department of Physiology School of Basic Medical Sciences Guangzhou Medical University Guangzhou China
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| |
Collapse
|
5
|
Feng W, Remedies CE, Obi IE, Aldous SR, Meera SI, Sanders PW, Inscho EW, Guan Z. Restoration of afferent arteriolar autoregulatory behavior in ischemia-reperfusion injury in rat kidneys. Am J Physiol Renal Physiol 2021; 320:F429-F441. [PMID: 33491564 PMCID: PMC7988813 DOI: 10.1152/ajprenal.00500.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/23/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Renal autoregulation is critical in maintaining stable renal blood flow (RBF) and glomerular filtration rate (GFR). Renal ischemia-reperfusion (IR)-induced kidney injury is characterized by reduced RBF and GFR. The mechanisms contributing to renal microvascular dysfunction in IR have not been fully determined. We hypothesized that increased reactive oxygen species (ROS) contributed to impaired renal autoregulatory capability in IR rats. Afferent arteriolar autoregulatory behavior was assessed using the blood-perfused juxtamedullary nephron preparation. IR was induced by 60 min of bilateral renal artery occlusion followed by 24 h of reperfusion. Afferent arterioles from sham rats exhibited normal autoregulatory behavior. Stepwise increases in perfusion pressure caused pressure-dependent vasoconstriction to 65 ± 3% of baseline diameter (13.2 ± 0.4 μm) at 170 mmHg. In contrast, pressure-mediated vasoconstriction was markedly attenuated in IR rats. Baseline diameter averaged 11.7 ± 0.5 µm and remained between 90% and 101% of baseline over 65-170 mmHg, indicating impaired autoregulatory function. Acute antioxidant administration (tempol or apocynin) to IR kidneys for 20 min increased baseline diameter and improved autoregulatory capability, such that the pressure-diameter profiles were indistinguishable from those of sham kidneys. Furthermore, the addition of polyethylene glycol superoxide dismutase or polyethylene glycol-catalase to the perfusate blood also restored afferent arteriolar autoregulatory responsiveness in IR rats, indicating the involvement of superoxide and/or hydrogen peroxide. IR elevated mRNA expression of NADPH oxidase subunits and monocyte chemoattractant protein-1 in renal tissue homogenates, and this was prevented by tempol pretreatment. These results suggest that ROS accumulation, likely involving superoxide and/or hydrogen peroxide, impairs renal autoregulation in IR rats in a reversible fashion.NEW & NOTEWORTHY Renal ischemia-reperfusion (IR) leads to renal microvascular dysfunction manifested by impaired afferent arteriolar autoregulatory efficiency. Acute administration of scavengers of reactive oxygen species, polyethylene glycol-superoxide dismutase, or polyethylene glycol-catalase following renal IR restored afferent arteriolar autoregulatory capability in IR rats, indicating that renal IR led to reversible impairment of afferent arteriolar autoregulatory capability. Intervention with antioxidant treatment following IR may improve outcomes in patients by preserving renovascular autoregulatory function and potentially preventing the progression to chronic kidney disease after acute kidney injury.
Collapse
Affiliation(s)
- Wenguang Feng
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Colton E Remedies
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ijeoma E Obi
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephen R Aldous
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Samia I Meera
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Edward W Inscho
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhengrong Guan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
6
|
Zhou S, Guo J, Zhao L, Liao Y, Zhou Q, Cui Y, Hu W, Chen J, Ren X, Wei Q, Jiang S, Zheng Y, Li L, Wilcox CS, Persson PB, Patzak A, Tian J, Yin Lai E. ADAMTS13 inhibits oxidative stress and ameliorates progressive chronic kidney disease following ischaemia/reperfusion injury. Acta Physiol (Oxf) 2021; 231:e13586. [PMID: 33226724 DOI: 10.1111/apha.13586] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/27/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022]
Abstract
AIMS Reduced A Disintegrin And Metalloproteinase with a ThromboSpondin type 1 motif member 13 (ADAMTS13) levels are observed in kidney disease. We test whether recombinant human ADAMTS13 (rhADAMTS13) mitigates renal injury in chronic kidney disease (CKD) and the potential mechanisms. METHODS CKD was established 3 months after ischaemia/reperfusion (IR). ADAMTS13 and von Willebrand factor (vWF) levels, renal function and morphological changes were analysed. Afferent arteriolar responses to angiotensin II (Ang II) and acetylcholine (ACh) were measured. Oxidative stress-related molecules were detected. RESULTS Higher vWF and lower ADAMTS13 levels were observed in CKD mice, which were markedly attenuated by rhADAMTS13. rhADAMTS13 alleviated renal dysfunction, as documented by decreased blood urea nitrogen (BUN), serum creatinine, kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) levels in CKD mice. Moreover, rhADAMTS13 attenuated transforming growth factor (TGF)-β1/Smad3 activation. Plasma vWF: ADAMTS13 ratio showed positive correlations with malondialdehyde (MDA), hydrogen peroxide (H2 O2 ) and proteinuria, and correlated inversely with superoxide dismutase (SOD) and catalase (CAT). Finally, rhADAMTS13 inhibited reactive oxygen species (ROS) levels and improved microvascular functional disorders, accompanied by the inhibition of glycogen synthase kinase (GSK) 3β hyperactivity and upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) expression. CONCLUSIONS Acute kidney injury (AKI) reduces the expression of ADAMTS13 that contributes to progressive CKD, microvascular dysfunction, oxidative stress, inhibition of Nrf2 activity and renal histopathological damage. All of which can be alleviated by administration of rhADAMTS13.
Collapse
Affiliation(s)
- Suhan Zhou
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Jie Guo
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Liang Zhao
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Department of Physiology School of Basic Medical Sciences Guangzhou Medical University Guangzhou China
| | - Yixin Liao
- Department of Obstetrics and Gynecology Nanfang HospitalSouthern Medical University Guangzhou China
| | - Qin Zhou
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Yu Cui
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Weipeng Hu
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Jianghua Chen
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Xiaoqiu Ren
- Department of Radiation Oncology Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Qichun Wei
- Department of Radiation Oncology Second Affiliated HospitalZhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - Yali Zheng
- Department of Nephrology Ningxia people’s hospital Yinchuan China
| | - Lingli Li
- Division of Nephrology and Hypertension, and Hypertension Research Center Georgetown University Washington DC USA
| | - Christopher S. Wilcox
- Division of Nephrology and Hypertension, and Hypertension Research Center Georgetown University Washington DC USA
| | - Pontus B. Persson
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Andreas Patzak
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Jiong Tian
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
| | - En Yin Lai
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences Zhejiang University School of Medicine Hangzhou China
- Institute of Vegetative Physiology Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
- Department of Physiology School of Basic Medical Sciences Guangzhou Medical University Guangzhou China
| |
Collapse
|
7
|
Zhao L, Cao X, Li L, Wang Q, Zhou S, Xu N, Jiang S, Chen L, Schmidt MO, Wei Q, Zhao J, Labes R, Patzak A, Wilcox CS, Fu X, Wellstein A, Lai EY. Acute Kidney Injury Sensitizes the Brain Vasculature to Ang II (Angiotensin II) Constriction via FGFBP1 (Fibroblast Growth Factor Binding Protein 1). Hypertension 2020; 76:1924-1934. [PMID: 33040621 PMCID: PMC9112323 DOI: 10.1161/hypertensionaha.120.15582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/14/2020] [Indexed: 12/26/2022]
Abstract
Acute kidney injury (AKI) causes multiple organ dysfunction. Here, we identify a possible mechanism that can drive brain vessel injury after AKI. We induced 30-minute bilateral renal ischemia-reperfusion injury in C57Bl/6 mice and isolated brain microvessels and macrovessels 24 hours or 1 week later to test their responses to vasoconstrictors and found that after AKI brain vessels were sensitized to Ang II (angiotensin II). Upregulation of FGF2 (fibroblast growth factor 2) and FGFBP1 (FGF binding protein 1) expression in both serum and kidney tissue after AKI suggested a potential contribution to the vascular sensitization. Administration of FGF2 and FGFBP1 proteins to isolated healthy brain vessels mimicked the sensitization to Ang II after AKI. Brain vessels in Fgfbp1-/- AKI mice failed to induce Ang II sensitization. Complementary to this, systemic treatment with the clinically used FGF receptor kinase inhibitor BGJ398 (Infigratinib) reversed the AKI-induced brain vascular sensitization to Ang II. All these findings lead to the conclusion that FGFBP1 is especially necessary for AKI-mediated brain vascular sensitization to Ang II and inhibitors of FGFR pathway may be beneficial in preventing AKI-induced brain vessel injury.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
- Institute of Vegetative Physiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
| | - Xiaoyun Cao
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lingli Li
- Division of Nephrology and Hypertension, Georgetown University, Washington, DC 20007, USA
| | - Qin Wang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Suhan Zhou
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Nan Xu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shan Jiang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Limeng Chen
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Marcel O. Schmidt
- Lombardi Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Qichun Wei
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jingwei Zhao
- Department of Anatomy, Histology and Embryology, Institute of Neuroscience, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Robert Labes
- Institute of Vegetative Physiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
| | - Andreas Patzak
- Institute of Vegetative Physiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
| | - Christopher S. Wilcox
- Division of Nephrology and Hypertension, Georgetown University, Washington, DC 20007, USA
| | - Xiaodong Fu
- Department of Gynecology and Obstetrics, the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511518, China
| | - Anton Wellstein
- Lombardi Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - En Yin Lai
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310003, China
- Institute of Vegetative Physiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
- Division of Nephrology and Hypertension, Georgetown University, Washington, DC 20007, USA
| |
Collapse
|
8
|
Xu N, Jiang S, Persson PB, Persson EAG, Lai EY, Patzak A. Reactive oxygen species in renal vascular function. Acta Physiol (Oxf) 2020; 229:e13477. [PMID: 32311827 DOI: 10.1111/apha.13477] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are produced by the aerobic metabolism. The imbalance between production of ROS and antioxidant defence in any cell compartment is associated with cell damage and may play an important role in the pathogenesis of renal disease. NADPH oxidase (NOX) family is the major ROS source in the vasculature and modulates renal perfusion. Upregulation of Ang II and adenosine activates NOX via AT1R and A1R in renal microvessels, leading to superoxide production. Oxidative stress in the kidney prompts renal vascular remodelling and increases preglomerular resistance. These are key elements in hypertension, acute and chronic kidney injury, as well as diabetic nephropathy. Renal afferent arterioles (Af), the primary resistance vessel in the kidney, fine tune renal hemodynamics and impact on blood pressure. Vice versa, ROS increase hypertension and diabetes, resulting in upregulation of Af vasoconstriction, enhancement of myogenic responses and change of tubuloglomerular feedback (TGF), which further promotes hypertension and diabetic nephropathy. In the following, we highlight oxidative stress in the function and dysfunction of renal hemodynamics. The renal microcirculatory alterations brought about by ROS importantly contribute to the pathophysiology of kidney injury, hypertension and diabetes.
Collapse
Affiliation(s)
- Nan Xu
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Pontus B. Persson
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | | | - En Yin Lai
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Andreas Patzak
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
9
|
Zhan H, Han P, Wang M, Wang Y, Weng W, Yu X, Yuan C, Li Y, Shao M, Sun H. Combination of astragaloside IV and ACEi ameliorates renal injuries in db/db mice. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:827-836. [PMID: 32509053 PMCID: PMC7270657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/26/2020] [Indexed: 06/11/2023]
Abstract
Evidences demonstrated that the effect on anti-proteinuria and renal protection of Chinese herbs combination with ACEi or ARB seemed to be better than ACEi or ARB alone. Astragaloside IV could decrease the urinary albumin excretion rate and could protect against renal injuries linking to its anti-oxidation ability. We aimed to investigate the effect of astragaloside IV combined with ACEi on diabetic nephropathy and to explore whether its underlying mechanism is dependent on anti-oxidation. 8-week-old male experiment mice were randomly assigned to five groups: lean wild type (wt) group, db/db group, db/db + astragaloside IV group, db/db + enalapril group, db/db + combination therapy with astragaloside IV and enalapril group. During the experiment, 24 hours urinary albumin, fasting glucose, body weight, and metabolic parameters were monitored in regular intervals. At the end of the study, tail blood pressure, serum H2O2, lipid, and liver function were measured and kidney histological injuries were evaluated. Results of the study indicated that combination therapy with astragaloside IV and ACEi further reduced 24 hours urinary albumin excretion rate, blood pressure, and body weight. Combination therapy reduced the foot process width, glomerular base membrane thickness, glomerular tuft cell proliferation, tubular cell atrophy, tubular base membrane thickness, and improved tubular cell proliferation. It modulated the body H2O2 metabolism and up-regulated the expression of the catalase in renal cortex. Astragaloside IV combined with ACEi exerted renal protective effects in db/db mice more significantly than their individual used. The mechanism possibly involved their synergistic effects on anti-oxidation.
Collapse
Affiliation(s)
- Hongyue Zhan
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
- Department of Critical Care Medicine, Shantou Hospital of Traditional Chinese MedicineShantou, Guangdong, China
| | - Pengxun Han
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Menghua Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Yao Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Wenci Weng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Xuewen Yu
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Changjian Yuan
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Yuyan Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Mumin Shao
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| | - Huili Sun
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, Guangdong, China
| |
Collapse
|
10
|
Jiang S, Wang X, Wei J, Zhang G, Zhang J, Xie P, Xu L, Wang L, Zhao L, Li L, Wilcox CS, Chen J, Lai EY, Liu R. NaHCO 3 Dilates Mouse Afferent Arteriole Via Na +/HCO 3- Cotransporters NBCs. Hypertension 2019; 74:1104-1112. [PMID: 31522618 DOI: 10.1161/hypertensionaha.119.13235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sodium bicarbonate has long been used to treat chronic kidney disease. It has been demonstrated to slow the decline in glomerular filtration rate in chronic kidney disease patient; however, the mechanisms are not completely understood. We hypothesized that NaHCO3 dilates afferent arterioles (Af-Art) by stimulating nitric oxide (NO) release mediated by the Na+/HCO3- cotransporter (NBC) contributing to the elevation in glomerular filtration rate. Isolated microperfused mouse renal Af-Art, preconstricted with norepinephrine (1 µmol/L), dilated 45±2% (n=6, P<0.05) in response to NaHCO3 (44 mmol/L). Whereas, NaCl solution containing the same Na+ concentration was not effective. The mRNA for NBCn1 and NBCe1 were detected in microdissected Af-Art using reverse transcription-polymerase chain reaction and quantitative polymerase chain reaction. The Af-Art intracellular pH measured with 2',7'-bis-(2-carboxyethyl)-5-(and-6) carboxyfluorescein, acetoxymethyl ester increased significantly by 0.29±0.02 (n=6; P<0.05) in the presence of NaHCO3, which was blunted by N-cyanosulphonamide compound (S0859) that is an inhibitor of the NBC family. After clamping the intracellular pH with 10 μM nigericin, changing the bath solution pH from 7.4 to 7.8 still dilates the Af-Art by 53±4% (n=7; P<0.005) and increases NO generation by 22±3% (n=7; P<0.005). Both pH-induced NO generation and vasodilation were blocked by L-NG-Nitroarginine Methyl Ester. NaHCO3 increased NO generation in Af-Art by 19±4% (n=5; P<0.005) and elevated glomerular filtration rate in conscious mice by 36% (233 versus 318 ul/min; n=9-10; P<0.0001). S0859 and L-NG-nitroarginine methyl ester blocked NaHCO3-induced increases in NO generation and vasodilation. We conclude that NBCn1 and NBCe1 are expressed in Af-Art and that NaHCO3 dilates Af-Art via NBCs mediated by NO that increases the glomerular filtration rate.
Collapse
Affiliation(s)
- Shan Jiang
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.).,Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.)
| | - Ximing Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.).,Shandong Provincial Hospital, Affiliated Hospital of Shandong University, Jinan, China (X.W.)
| | - Jin Wei
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.)
| | - Gensheng Zhang
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.).,Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.)
| | - Jie Zhang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.)
| | - Peng Xie
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.)
| | - Lan Xu
- College of Public Health, University of South Florida, Tampa (L.X.)
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.)
| | - Liang Zhao
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.).,Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, China (L.Z., E.Y.L.).,Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Germany (L.Z., E.Y.L.)
| | - Lingli Li
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.).,Division of Nephrology and Hypertension, and Hypertension Center, Georgetown University, Washington, DC (L.L., C.S.W.)
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension, and Hypertension Center, Georgetown University, Washington, DC (L.L., C.S.W.)
| | - Jianghua Chen
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.)
| | - En Yin Lai
- From Kidney Disease Center, the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China (S.J., G.Z., P.X., L.Z., L.L., J.C., E.Y.L.).,Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, China (L.Z., E.Y.L.).,Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Germany (L.Z., E.Y.L.)
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (S.J., X.W., J.W., G.Z., J.Z., L.W., R.L.)
| |
Collapse
|
11
|
Zhou S, Jiang S, Guo J, Xu N, Wang Q, Zhang G, Zhao L, Zhou Q, Fu X, Li L, Patzak A, Hultström M, Lai EY. ADAMTS13 protects mice against renal ischemia-reperfusion injury by reducing inflammation and improving endothelial function. Am J Physiol Renal Physiol 2019; 316:F134-F145. [DOI: 10.1152/ajprenal.00405.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute kidney injury (AKI) is a serious condition without efficient therapeutic options. Recent studies have indicated that recombinant human a disintegrin and metalloprotease with thrombospondin motifs 13 (rhADAMTS13) provides protection against inflammation. Therefore, we hypothesized that ADAMTS13 might protect against AKI by reducing inflammation. Bilateral renal ischemia-reperfusion injury (I/R) was used as AKI models in this study. Prophylactic infusion of rhADAMTS13 was employed to investigate potential mechanisms of renal protection. Renal function, inflammation, and microvascular endothelial function were assessed after 24 h of reperfusion. Our results showed that I/R mice increased plasma von Willebrand factor levels but decreased ADAMTS13 expression. Administration of rhADAMTS13 to I/R mice recovered renal function, histological injury, and apoptosis. Renal inflammation was reduced by rhADAMTS13, accompanied with the downregulation of p38/extracellular signal-regulated protein kinase phosphorylation and cyclooxygenase-2 expression. rhADAMTS13 restored vasodilation in afferent arterioles in I/R mice. Furthermore, rhADAMTS13 treatment enhanced phosphorylation of Akt at Ser473 and eNOS at Ser1177. Administration of the Akt pathway inhibitor wortmannin reduced the protective effect of rhADAMTS13. Our conclusions are that treatment with rhADAMTS13 ameliorates renal I/R injury by reducing inflammation, tubular cell apoptosis, and improving microvascular endothelial dysfunction. rhADAMTS13 could be a promising strategy to treat AKI in clinical settings.
Collapse
Affiliation(s)
- Suhan Zhou
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Jiang
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Guo
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan Xu
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Wang
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Gensheng Zhang
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Zhao
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Institute of Vegetative Physiology, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Qin Zhou
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Fu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lingli Li
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, District of Columbia
| | - Andreas Patzak
- Institute of Vegetative Physiology, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Hultström
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - En Yin Lai
- Kidney Disease Center of First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, District of Columbia
| |
Collapse
|
12
|
Westphal A, Mrowka R. New insights into the astonishing diversity of hormone functions. Acta Physiol (Oxf) 2018; 224:e13188. [PMID: 30240073 DOI: 10.1111/apha.13188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 09/17/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Anika Westphal
- Klinik für Innere Medizin III, AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| | - Ralf Mrowka
- Klinik für Innere Medizin III, AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| |
Collapse
|
13
|
Li L, Lai EY, Luo Z, Solis G, Mendonca M, Griendling KK, Wellstein A, Welch WJ, Wilcox CS. High Salt Enhances Reactive Oxygen Species and Angiotensin II Contractions of Glomerular Afferent Arterioles From Mice With Reduced Renal Mass. Hypertension 2018; 72:1208-1216. [PMID: 30354808 PMCID: PMC6221452 DOI: 10.1161/hypertensionaha.118.11354] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022]
Abstract
High salt, Ang II (angiotensin II), and reactive oxygen species enhance progression of chronic kidney disease. We tested the hypothesis that a high salt intake generates specific reactive oxygen species to enhance Ang II contractions of afferent arterioles from mice with reduced renal mass (RRM). C57BL/6 mice were subjected to surgical RRM or sham operations and received 6% or 0.4% NaCl salt diet for 3 months. Ang II contractions were measured in perfused afferent arterioles and superoxide (O2-) and hydrogen peroxide (H2O2) by fluorescence microscopy. RRM enhanced the afferent arteriolar gene expression for p47phox and neutrophil oxidase (NOX) 2 and high salt intake in RRM mice enhanced gene expression for angiotensin type 1 receptors, POLDIP2 and NOX4 and reduced catalase. High salt in mice with RRM enhanced arteriolar O2- and H2O2 generation and maximal contractions to Ang II (10-6 mol/L) that were dependent on O2- because they were prevented by gene deletion of p47phox and on H2O2 because they were prevented by transgenic smooth muscle cell expression of catalase (tgCAT-SMC) and POLDIP2 gene deletion. Three months of tempol normalized arteriolar reactive oxygen species and Ang II contractions. However, arteriolar contractions to lower concentrations of Ang II (10-8 to 10-11 mol/L) were paradoxically inhibited by H2O2 and POLDIP2. In conclusion, both O2- from p47phox/NOX2 and H2O2 from NOX4/POLDIP2 enhance maximal arteriolar Ang II contractions from RRM mice during high salt, but H2O2 and NOX4/POLDIP2 reduce the sensitivity to lower concentrations of Ang II by >100-fold. Tempol prevents all of these changes in function.
Collapse
Affiliation(s)
- Lingli Li
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
| | - En Yin Lai
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou China
| | - Zaiming Luo
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
| | - Glenn Solis
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
| | - Margarida Mendonca
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
| | - Kathy K. Griendling
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta
| | - Anton Wellstein
- Lombardi Cancer Center, Georgetown University, Washington DC
| | - William J. Welch
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
| | - Christopher S. Wilcox
- Division of Nephrology and Hypertension, and Hypertension Research Center, Georgetown University, Washington, DC
| |
Collapse
|
14
|
Affiliation(s)
- Pontus B. Persson
- Charité - Universitätsmedizin Berlin; Corporate Member of Freie Universität Berlin; Humboldt-Universität zu Berlin; and Berlin Institute of Health; Institute of Vegetative Physiology; Berlin Germany
- Charité - Universitätsmedizin Berlin; Corporate Member of Freie Universität Berlin; Humboldt-Universität zu Berlin; and Berlin Institute of Health; Berlin Germany
| | - Anja Bondke Persson
- Charité - Universitätsmedizin Berlin; Corporate Member of Freie Universität Berlin; Humboldt-Universität zu Berlin; and Berlin Institute of Health; Institute of Vegetative Physiology; Berlin Germany
- Charité - Universitätsmedizin Berlin; Corporate Member of Freie Universität Berlin; Humboldt-Universität zu Berlin; and Berlin Institute of Health; Berlin Germany
| |
Collapse
|
15
|
Xu N, Wang Q, Jiang S, Wang Q, Hu W, Zhou S, Zhao L, Xie L, Chen J, Wellstein A, Lai EY. Fenofibrate improves vascular endothelial function and contractility in diabetic mice. Redox Biol 2018; 20:87-97. [PMID: 30296701 PMCID: PMC6174921 DOI: 10.1016/j.redox.2018.09.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/11/2018] [Accepted: 09/27/2018] [Indexed: 11/25/2022] Open
Abstract
Fenofibrate, a peroxisome proliferator-activated receptors α (PPARα) agonist, reduces vascular complications of diabetic patients but its protective mechanisms are not fully understood. Here we tested the hypothesis that fenofibrate improves vascular endothelial dysfunction by balancing endothelium-dependent relaxation and contractility of the aorta in diabetes mellitus (DM). In streptozotocin-induced diabetic mice, eight weeks of fenofibrate treatment (100 mg/Kg/d) improved endothelium dependent relaxation in the macro- and microvessels, increased nitric oxide (NO) levels, reduced renal damage markers and effects of the vasoconstrictor prostaglandin. Levels of superoxide dismutase and catalase were both reduced and hydrogen peroxide was increased in vehicle-treated DM, but these changes were reversed by fenofibrate treatment. Vasodilation of the aorta after fenofibrate treatment was reversed by PPARα or AMPKα inhibitors. Western blots showed that fenofibrate treatment elevated PPARα expression, induced liver kinase B1 (LKB1) translocation from the nucleus to the cytoplasm and activated AMP-activated protein kinase-α (AMPKα), thus activating endothelial NO synthase (eNOS). Also, fenofibrate treatment decreased NF-κB p65 and cyclooxygenase 2 proteins in aortas. Finally, incubation with indomethacin in vitro improved aortic contractility in diabetic mice. Overall, our results show that fenofibrate treatment in diabetic mice normalizes endothelial function by balancing vascular reactivity via increasing NO production and suppressing the vasoconstrictor prostaglandin, suggesting mechanism of action of fenofibrate in mediating diabetic vascular complications. Fenofibrate improves diabetic endothelial function is via PPAR/LKB1/AMPK/eNOS signal. Fenofibrate reduces diabetic endothelial contractility is via NF-κB/COX-2 pathway. Diabetes-associated oxidative stress is attenuated by fenofibrate treatment.
Collapse
Affiliation(s)
- Nan Xu
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qin Wang
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shan Jiang
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qijing Wang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weipeng Hu
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Suhan Zhou
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liang Zhao
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lanyu Xie
- Medical college, Nanchang University, Nanchang 330000, China
| | - Jianghua Chen
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Anton Wellstein
- Lombardi Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - En Yin Lai
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
16
|
Ackermann S, Mrowka R. Nephropathy: New aspects of mechanisms, diagnosis and therapy. Acta Physiol (Oxf) 2018; 224:e13162. [PMID: 29984559 DOI: 10.1111/apha.13162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Susanne Ackermann
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| | - Ralf Mrowka
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| |
Collapse
|
17
|
Allaqaband H, Gutterman DD, Kadlec AO. Physiological Consequences of Coronary Arteriolar Dysfunction and Its Influence on Cardiovascular Disease. Physiology (Bethesda) 2018; 33:338-347. [PMID: 30109826 PMCID: PMC6230549 DOI: 10.1152/physiol.00019.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
To date, the major focus of diagnostic modalities and interventions to treat coronary artery disease has been the large epicardial vessels. Despite substantial data showing that microcirculatory dysfunction is a strong predictor of future adverse cardiovascular events, very little research has gone into developing techniques for in vivo diagnosis and therapeutic interventions to improve microcirculatory function. In this review, we will discuss the pathophysiology of coronary arteriolar dysfunction, define its prognostic implications, evaluate the diagnostic modalities available, and provide speculation on current and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Hassan Allaqaband
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - David D Gutterman
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Medicine, Division of Cardiology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Veterans Administration Medical Center, Milwaukee, Wisconsin
| | - Andrew O Kadlec
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Medicine, Division of Cardiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
18
|
GRK2 knockdown in mice exacerbates kidney injury and alters renal mechanisms of blood pressure regulation. Sci Rep 2018; 8:11415. [PMID: 30061705 PMCID: PMC6065385 DOI: 10.1038/s41598-018-29876-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system regulates blood pressure and fluid balance in the body primarily via angiotensin receptor 1 (AT1R). Renal AT1R was found to be primarily responsible for Ang II-mediated hypertension. G protein-coupled receptor kinase 2 (GRK2) modulates AT1R desensitization and increased GRK2 protein expression is reported in hypertensive patients. However, the consequences of GRK2 inhibition on kidney functions remain unknown. We employed shGRK2 knockdown mice (shGRK2 mice) to test the role of GRK2 in kidney development and function that can be ultimately linked to the hypertensive phenotype detected in shGRK2 mice. GRK2 knockdown reduced kidney size, nephrogenesis and glomerular count, and impaired glomerular filtration. Glomerular damage in adult shGRK2 mice was associated with increased renin- and AT1R-mediated production of reactive oxygen species. The AT1R blocker, Losartan, normalized elevated blood pressure and markedly improved glomerular filtration in the shGRK2 knockdown mice. Our findings provide evidence for the crucial role of GRK2 in renal regulation of blood pressure. It also suggests that the detrimental outcomes of GRK2 inhibitors on the kidney should be carefully examined when used as antihypertensive.
Collapse
|
19
|
Zhang S, Huang Q, Wang Q, Wang Q, Cao X, Zhao L, Xu N, Zhuge Z, Mao J, Fu X, Liu R, Wilcox CS, Patzak A, Li L, Lai EY. Enhanced Renal Afferent Arteriolar Reactive Oxygen Species and Contractility to Endothelin-1 Are Associated with Canonical Wnt Signaling in Diabetic Mice. Kidney Blood Press Res 2018; 43:860-871. [PMID: 29870994 PMCID: PMC6050514 DOI: 10.1159/000490334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/24/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIMS Canonical Wnt signaling is involved in oxidative stress, vasculopathy and diabetes mellitus but its role in diabetic renal microvascular dysfunction is unclear. We tested the hypothesis that enhanced canonical Wnt signaling in renal afferent arterioles from diabetic mice increases reactive oxygen species (ROS) and contractions to endothelin-1 (ET-1). METHODS Streptozotocin-induced diabetes or control C57Bl/6 mice received vehicle or sulindac (40 mg·kg-1·day-1) to block Wnt signaling for 4 weeks. ET-1 contractions were measured by changes of afferent arteriolar diameter. Arteriolar H2O2, O2 -, protein expression and enzymatic activity were assessed using sensitive fluorescence probes, immunoblotting and colorimetric assay separately. RESULTS Compared to control, diabetic mouse afferent arteriole had increased O2- (+ 84%) and H2O2 (+ 91%) and enhanced responses to ET-1 at 10-8 mol·l-1 (-72±4% of versus -43±4%, P< 0.05) accompanied by reduced protein expressions and activities for catalase and superoxide dismutase 2 (SOD2). Arteriolar O2 - was increased further by ET-1 and contractions to ET-1 reduced by PEG-SOD in both groups whereas H2O2 unchanged by ET-1 and contractions were reduced by PEG-catalase selectively in diabetic mice. The Wnt signaling protein β-catenin was upregulated (3.3-fold decrease in p-β-catenin/β-catenin) while the glycogen synthase kinase-3β (GSK-3β) was downregulated (2.6-fold increase in p-GSK-3β/ GSK-3β) in preglomerular vessels of diabetic mice. Sulindac normalized the Wnt signaling proteins, arteriolar O2 -, H2O2 and ET-1 contractions while doubling microvascular catalase and SOD2 expression in diabetic mice. CONCLUSION Increased ROS, notably H2O2 contributes to enhanced afferent arteriolar responses to ET-1 in diabetes, which is closely associated with Wnt signaling. Antioxidant pharmacological strategies targeting Wnt signaling may improve vascular function in diabetic nephropathy.
Collapse
Affiliation(s)
- Suping Zhang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Huang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Physiology, Quanzhou Medical College, Quanzhou, China
| | - Qiaoling Wang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Wang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyun Cao
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Zhao
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Nan Xu
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengbing Zhuge
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Mao
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Fu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension, and Hypertension Center, Georgetown University, Washington, District of Columbia, USA
| | - Andreas Patzak
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lingli Li
- Division of Nephrology and Hypertension, and Hypertension Center, Georgetown University, Washington, District of Columbia, USA
| | - En Yin Lai
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China,
- Division of Nephrology and Hypertension, and Hypertension Center, Georgetown University, Washington, District of Columbia, USA,
| |
Collapse
|
20
|
Zhang G, Wang Q, Wang W, Yu M, Zhang S, Xu N, Zhou S, Cao X, Fu X, Ma Z, Liu R, Mao J, Lai EY. Tempol Protects Against Acute Renal Injury by Regulating PI3K/Akt/mTOR and GSK3β Signaling Cascades and Afferent Arteriolar Activity. Kidney Blood Press Res 2018; 43:904-913. [PMID: 29870982 PMCID: PMC6065105 DOI: 10.1159/000490338] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIMS Free radical scavenger tempol is a protective antioxidant against ischemic injury. Tubular epithelial apoptosis is one of the main changes in the renal ischemia/reperfusion (I/R) injury. Meanwhile some proteins related with apoptosis and inflammation are also involved in renal I/R injury. We tested the hypothesis that tempol protects against renal I/R injury by activating protein kinase B/mammalian target of rapamycin (PKB, Akt/mTOR) and glycogen synthase kinase 3β (GSK3β) pathways as well as the coordinating apoptosis and inflammation related proteins. METHODS The right renal pedicle of C57Bl/6 mouse was clamped for 30 minutes and the left kidney was removed in the study. The renal injury was assessed with serum parameters by an automatic chemistry analyzer. Renal expressions of Akt/mTOR and GSK3β pathways were measured by western blot in I/R mice treated with saline or tempol (50mg/kg) and compared with sham-operated mice. RESULTS The levels of blood urea nitrogen (BUN), creatinine and superoxide anion (O2.-) increased, and superoxide dismutase (SOD) and catalase (CAT) decreased significantly after renal I/R injury. However, tempol treatment prevented the changes. Besides, I/R injury reduced renal expression of p-Akt, p-GSK3β, p-mTOR, Bcl2 and increased NF-κB, p-JNK and p53 in kidney, tempol significantly normalized these changes. In addition, renal I/R injury reduced the response of afferent arteriole to Angiotensin II (Ang II), while tempol treatment improved the activity of afferent arteriole. CONCLUSION Tempol attenuates renal I/R injury. The protective mechanisms seem to relate with activation of PI3K/Akt/mTOR and GSK3β pathways, inhibition of cellular damage markers and inflammation factors, as well as improvement of afferent arteriolar activity.
Collapse
Affiliation(s)
- Gensheng Zhang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Wang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenwen Wang
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minghua Yu
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Suping Zhang
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan Xu
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Suhan Zhou
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyun Cao
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Fu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zufu Ma
- Department of Nephrology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Jianhua Mao
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - En Yin Lai
- Department of Physiology, and the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China,
| |
Collapse
|
21
|
Braun D, Zollbrecht C, Dietze S, Schubert R, Golz S, Summer H, Persson PB, Carlström M, Ludwig M, Patzak A. Hypoxia/Reoxygenation of Rat Renal Arteries Impairs Vasorelaxation via Modulation of Endothelium-Independent sGC/cGMP/PKG Signaling. Front Physiol 2018; 9:480. [PMID: 29773995 PMCID: PMC5943512 DOI: 10.3389/fphys.2018.00480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/16/2018] [Indexed: 11/13/2022] Open
Abstract
Ischemia/reperfusion injury holds a key position in many pathological conditions such as acute kidney injury and in the transition to chronic stages of renal damage. We hypothesized that besides a reported disproportional activation of vasoconstrictor response, hypoxia/reoxygenation (H/R) adversely affects endothelial dilatory systems and impairs relaxation in renal arteries. Rat renal interlobar arteries were studied under isometric conditions. Hypoxia was induced by application of 95% N2, 5% CO2 for 60 min to the bath solution, followed by a 10 min period of reoxygenation (95% O2, 5% CO2). The effect of H/R on relaxation was assessed using various inhibitors of endothelial dilatory systems. mRNA expression of phosphodiesterase 5 (PDE5), NADPH oxidases (NOX), and nitric oxide synthase (NOS) isoforms were determined using qRT-PCR; cGMP was assayed with direct cGMP ELISA. Acetylcholine induced relaxation was impaired after H/R. Inhibition of the NOS isoforms with L-NAME, and cyclooxygenases (COXs) by indomethacin did not abolish the H/R effect. Moreover, blocking the calcium activated potassium channels KCa3.1 and KCa2.1, the main mediators of the endothelium-derived hyperpolarizing factor, with TRAM34 and UCL1684, respectively, showed similar effects in H/R and control. Arterial stiffness did not differ comparing H/R with controls, indicating no impact of H/R on passive vessel properties. Moreover, superoxide was not responsible for the observed H/R effect. Remarkably, H/R attenuated the endothelium-independent relaxation by sodium nitroprusside, suggesting endothelium-independent mechanisms of H/R action. Investigating the signaling downstream of NO revealed significantly decreased cGMP and impaired relaxation during PDE5 inhibition with sildenafil after H/R. Inhibition of PKG, the target of cGMP, did not normalize SNP-induced relaxation following H/R. However, the soluble guanylyl cyclase (sGC) inhibitor ODQ abolished the H/R effect on relaxation. The mRNA expressions of the endothelial and the inducible NOS were reduced. NOX and PDE5 mRNA were similarly expressed in H/R and control. Our results provide new evidence that impaired renal artery relaxation after H/R is due to a dysregulation of sGC leading to decreased cGMP levels. The presented mechanism might contribute to an insufficient renal reperfusion after ischemia and should be considered in its pathophysiology.
Collapse
Affiliation(s)
- Diana Braun
- Renal Vessel Physiology Group, Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christa Zollbrecht
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Stefanie Dietze
- Renal Vessel Physiology Group, Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | - Pontus B Persson
- Renal Vessel Physiology Group, Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marion Ludwig
- Renal Vessel Physiology Group, Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Patzak
- Renal Vessel Physiology Group, Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Vegetative Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
22
|
Xu Y, Hu N, Jiang W, Yuan HF, Zheng DH. Curcumin-carrying nanoparticles prevent ischemia-reperfusion injury in human renal cells. Oncotarget 2018; 7:87390-87401. [PMID: 27901497 PMCID: PMC5349996 DOI: 10.18632/oncotarget.13626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a major complication in clinical practice. However, despite its frequency, effective preventive/treatment strategies for this condition are scarce. Curcumin possesses antioxidant properties and is a promising potential protective agent against renal IRI, but its poor water solubility restricts its application. In this study, we constructed curcumin-carrying distearoylphosphatidylethanolamine-polyethylene glycol nanoparticles (Cur-NPs), and their effect on HK-2 cells exposed to IRI was examined in vitro. Curcumin encapsulated in NPs demonstrated improved water solubility and slowed release. Compared with the IRI and Curcumin groups, Cur-NP groups displayed significantly improved cell viability, downregulated protein expression levels of caspase-3 and Bax, upregulated expression of Bcl-2 protein, increased antioxidant superoxide dismutase level, and reduced apoptotic rate, reactive oxygen species level, and malondialdehyde content. Results clearly showed that Cur-NPs demonstrated good water solubility and slow release, as well as exerted protective effects against oxidative stress in cultured HK-2 cells exposed to IRI.
Collapse
Affiliation(s)
- Yong Xu
- Department of Nephrology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an 223002, China
| | - Ning Hu
- Department of Nephrology, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, China
| | - Wei Jiang
- Department of Nephrology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an 223002, China
| | - Hong-Fang Yuan
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dong-Hui Zheng
- Department of Nephrology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an 223002, China
| |
Collapse
|
23
|
Shaterzadeh-Yazdi H, Noorbakhsh MF, Samarghandian S, Farkhondeh T. An Overview on Renoprotective Effects of Thymoquinone. KIDNEY DISEASES 2018; 4:74-82. [PMID: 29998122 DOI: 10.1159/000486829] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022]
Abstract
Background Kidneys as vital organs remove waste material from blood. Additionally, they may also have a role in the electrolyte balance, regulation of blood pressure, and red blood cell genesis. Kidney diseases may be caused by several factors such as ischemia/reperfusion injury, diabetes, and nephrotoxic agents. Oxidative stress and inflammation are involved in the pathogenesis and progression of kidney diseases. Traditionally, natural antioxidants are used for treatment of renal failure in various countries. Summary People usually select natural antioxidants since they have an opinion that herbal medicine has not any important side effects. Nigella sativa is a flavoring herb that is widely used as a condiment and as a remedy for many disorders. Thymoquinone (TQ), the most important component of black seeds, mainly oil, is considered as an active agent responsible for a lot of the seed's useful effects. This review describes the protective roles and related mechanisms of TQ against renal disorders. The search terms, including TQ, antioxidant, renal ischemia-reperfusion, diabetic nephropathy, and nephrotoxic agent were searched in scientific databases. TQ showed anti-inflammatory and antioxidant properties in animal and in vitro models of several renal diseases caused by inflammation and oxidative stress. Key Messages Experimental studies have shown beneficial effects of TQ against renal diseases; however, well-designed clinical trials in humans are required to confirm these effects.
Collapse
Affiliation(s)
- Hanieh Shaterzadeh-Yazdi
- Research Center of Pardis Hospital, Mashhad, Iran.,Nastaran Center for Cancer Prevention, Mashhad, Iran
| | - Mohammad-Foad Noorbakhsh
- Division of Pharmacology and Toxicology, Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
24
|
Hultström M, Becirovic-Agic M, Jönsson S. Comparison of acute kidney injury of different etiology reveals in-common mechanisms of tissue damage. Physiol Genomics 2017; 50:127-141. [PMID: 29341864 DOI: 10.1152/physiolgenomics.00037.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) is a syndrome of reduced glomerular filtration rate and urine production caused by a number of different diseases. It is associated with renal tissue damage. This tissue damage can cause tubular atrophy and interstitial fibrosis that leads to nephron loss and progression of chronic kidney disease (CKD). This review describes the in-common mechanisms behind tissue damage in AKI caused by different underlying diseases. Comparing six high-quality microarray studies of renal gene expression after AKI in disease models (gram-negative sepsis, gram-positive sepsis, ischemia-reperfusion, malignant hypertension, rhabdomyolysis, and cisplatin toxicity) identified 5,254 differentially expressed genes in at least one of the AKI models; 66% of genes were found only in one model, showing that there are unique features to AKI depending on the underlying disease. There were in-common features in the form of four genes that were differentially expressed in all six models, 49 in at least five, and 215 were found in common between at least four models. Gene ontology enrichment analysis could be broadly categorized into the injurious processes hypoxia, oxidative stress, and inflammation, as well as the cellular outcomes of cell death and tissue remodeling in the form of epithelial-to-mesenchymal transition. Pathway analysis showed that MYC is a central connection in the network of activated genes in-common to AKI, which suggests that it may be a central regulator of renal gene expression in tissue injury during AKI. The outlining of this molecular network may be useful for understanding progression from AKI to CKD.
Collapse
Affiliation(s)
- Michael Hultström
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University , Uppsala , Sweden.,Anaesthesia and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University , Uppsala , Sweden
| | - Mediha Becirovic-Agic
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University , Uppsala , Sweden
| | - Sofia Jönsson
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University , Uppsala , Sweden
| |
Collapse
|
25
|
Affiliation(s)
- S. Reuter
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| | - R. Mrowka
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| |
Collapse
|
26
|
Affiliation(s)
- R. Mrowka
- Klinik fuer Innere Medizin III; AG Experimentelle Nephrologie; Universitaetsklinikum Jena; Jena Germany
| |
Collapse
|