1
|
Zheng Z, Hu W, Ji C, Zhang X, Ding X, Zhou S, Li J, Chen G. A study of the difference in biochemical metabolism between patients with unilateral and bilateral upper urinary tract stones. Sci Rep 2024; 14:30154. [PMID: 39627370 PMCID: PMC11615316 DOI: 10.1038/s41598-024-81454-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
Bilateral upper urinary tract stones are more likely to lead to impairment of renal function, but few biochemical metabolic studies of bilateral upper urinary tract stones have been reported. We collected clinical data from 555 patients with upper urinary tract stones admitted to Beijing Tsinghua Changgung Hospital from June 2020 to June 2024, and divided them into unilateral and bilateral stone groups by CT scans, analysed the metabolic differences between unilateral and bilateral stone groups by statistical methods, and used multifactorial logistic regression analysis to explore the risk factors that might affect the formation of bilateral stones. A total of 281 cases of unilateral and 274 cases of bilateral stones were identified. The proportion of male patients in the bilateral group was higher than that in the unilateral group (P < 0.05). The most prevalent major stone component was calcium oxalate monohydrate (48.1%), with a significantly higher prevalence of cystine stones observed in the bilateral stone group (1.8%) compared to the unilateral stone group (0.4%) (P < 0.05). Blood uric acid, blood BUN, blood creatinine, urine pH, and 24-hour urine output were higher in the bilateral stone group than in the unilateral group (P < 0.05). The most prevalent metabolic abnormality was low urine volume (45.7%). Bilateral stone group had higher proportion of patients with hyperuricemia (P < 0.05). The results of the multivariate logistic regression analysis showed that male gender (OR 1.489, 95% CI 1.028-2.157) and hyperuricemia (OR 1.662, 95% CI 1.113-2.482) were associated with an increased risk of bilateral stone formation (P < 0.05). There are significant differences in biochemical metabolism between unilateral and bilateral upper urinary tract stones. The most common metabolic abnormality in patients with urolithiasis is low urine output, and aggressive water intake is effective in preventing stone formation. For patients with hyperuricemia, a strict dietary regimen is imperative to mitigate the likelihood of bilateral stone formation.
Collapse
Affiliation(s)
- Zhibin Zheng
- Department of Urology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Department of Urology, Qinghai University Affiliated Hospital, Xining, 810000, China
| | - Weiguo Hu
- Department of Urology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| | - Chaoyue Ji
- Department of Urology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xuming Zhang
- Department of Urology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xijie Ding
- Department of Urology, Qinghai University Affiliated Hospital, Xining, 810000, China
| | - Shaobo Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Xining, 810000, China
| | - Jianxing Li
- Department of Urology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Guojun Chen
- Department of Urology, Qinghai University Affiliated Hospital, Xining, 810000, China
| |
Collapse
|
2
|
Golmohammadi M, Ivraghi MS, Hasan EK, Huldani H, Zamanian MY, Rouzbahani S, Mustafa YF, Al-Hasnawi SS, Alazbjee AAA, Khalajimoqim F, Khalaj F. Protective effects of pioglitazone in renal ischemia-reperfusion injury (RIRI): focus on oxidative stress and inflammation. Clin Exp Nephrol 2024; 28:955-968. [PMID: 38935212 DOI: 10.1007/s10157-024-02525-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (RIRI) is a critical phenomenon that compromises renal function and is the most serious health concern related to acute kidney injury (AKI). Pioglitazone (Pio) is a known agonist of peroxisome proliferator-activated receptor-gamma (PPAR-γ). PPAR-γ is a nuclear receptor that regulates genes involved in inflammation, metabolism, and cellular differentiation. Activation of PPAR-γ is associated with antiinflammatory and antioxidant effects, which are relevant to the pathophysiology of RIRI. This study aimed to investigate the protective effects of Pio in RIRI, focusing on oxidative stress and inflammation. METHODS We conducted a comprehensive literature search using electronic databases, including PubMed, ScienceDirect, Web of Science, Scopus, and Google Scholar. RESULTS The results of this study demonstrated that Pio has antioxidant, anti-inflammatory, and anti-apoptotic activities that counteract the consequences of RIRI. The study also discussed the underlying mechanisms, including the modulation of various pathways such as TNF-α, NF-κB signaling systems, STAT3 pathway, KIM-1 and NGAL pathways, AMPK phosphorylation, and autophagy flux. Additionally, the study presented a summary of various animal studies that support the potential protective effects of Pio in RIRI. CONCLUSION Our findings suggest that Pio could protect the kidneys from RIRI by improving antioxidant capacity and decreasing inflammation. Therefore, these findings support the potential of Pio as a therapeutic strategy for preventing RIRI in different clinical conditions.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1988873554, Iran
| | | | | | - Huldani Huldani
- Department of Physiology, Faculty of Medicine Lambung, Mangkurat University, South Kalimantan, Banjarmasin, Indonesia
| | - Mohammad Yasin Zamanian
- Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Shiva Rouzbahani
- Miller School of Medicine, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
- Department of Community Medicine and Family Physician, School of Medicine, Isfahan University of Medical Sciences, Hezar Jarib Blvd, Isfahan, Iran
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | | | - Faranak Khalajimoqim
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Fattaneh Khalaj
- Digestive Diseases Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Shirzad H, Mousavinezhad SA, Panji M, Ala M. Amlodipine alleviates renal ischemia/reperfusion injury in rats through Nrf2/Sestrin2/PGC-1α/TFAM Pathway. BMC Pharmacol Toxicol 2023; 24:82. [PMID: 38129888 PMCID: PMC10740300 DOI: 10.1186/s40360-023-00722-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Previously, observational studies showed that amlodipine can mitigate calcineurin inhibitor- and contrast-induced acute kidney injury (AKI). Herein, we aimed to measure the effect of amlodipine on renal ischemia/reperfusion (I/R) injury and find the underlying mechanisms. MATERIALS AND METHODS Bilateral renal I/R was induced by clamping the hilum of both kidneys for 30 min. The first dose of amlodipine 10 mg/kg was gavaged before anesthesia. The second dose of amlodipine was administered 24 h after the first dose. Forty-eight hours after I/R, rats were anesthetized, and their blood and tissue specimens were collected. RESULTS Amlodipine significantly decreased the elevated serum levels of creatinine and blood urea nitrogen (BUN) and mitigated tissue damage in hematoxylin & eosin (H&E) staining. Amlodipine strongly reduced the tissue levels of malondialdehyde (MDA), interleukin 1β (IL1β), and tumor necrosis factor α (TNF-α). Amlodipine enhanced antioxidant defense by upregulating nuclear factor erythroid 2-related factor 2 (Nrf2) and Sestrin2. Furthermore, amlodipine significantly improved mitochondrial biogenesis by promoting Sestrin2/peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α)/mitochondrial transcription factor A (TFAM) pathway. It also enhanced autophagy and attenuated apoptosis, evidenced by increased LC3-II/LC3-I and bcl2/bax ratios after renal I/R. CONCLUSION These findings suggest that amlodipine protects against renal I/R through Nrf2/Sestrin2/PGC-1α/TFAM Pathway.
Collapse
Affiliation(s)
- Hadi Shirzad
- Research Center for Life, Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarters, Tehran, Iran
| | - Seyed Amin Mousavinezhad
- Research Center for Life, Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarters, Tehran, Iran
| | - Mohammad Panji
- Research Center for Life, Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarters, Tehran, Iran
| | - Moin Ala
- Research Center for Life, Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarters, Tehran, Iran.
| |
Collapse
|
4
|
Zhang C, Guan G, Wang J, Wei H, Cai J. MicroRNA-192-5p downregulates Fat Mass and Obesity-associated Protein to aggravate renal ischemia/reperfusion injury. Ren Fail 2023; 45:2285869. [PMID: 38044851 PMCID: PMC11001322 DOI: 10.1080/0886022x.2023.2285869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/15/2023] [Indexed: 12/05/2023] Open
Abstract
Acute kidney injury (AKI) is a common disorder without effective therapy yet. Renal ischemia/reperfusion (I/R) injury is a common cause of AKI. MicroRNA miR-192-5p has been previously reported to be upregulated in AKI models. However, its functional role in renal I/R injury is not fully understood. This study aimed to investigate the effects and the underlying mechanism of miR-192-5p in renal I/R progression. Hypoxia/reoxygenation (H/R)-induced cell injury model in HK-2 cells and I/R-induced renal injury model in mice were established in this study. Cell counting kit-8 assay was performed to determine cell viability. Quantitative real-time PCR and western blot analysis were performed to detect gene expressions. Hematoxylin-eosin and periodic acid-Schiff staining were performed to observe the histopathological changes. Enzyme-linked immunosorbent assay was performed to detect the kidney markers' expression. In vivo and in vitro results showed that miR-192-5p was up-regulated in the I/R-induced mice model and H/R-induced cell model, and miR-192-5p overexpression exacerbated I/R-induced renal damage. Then, the downstream target of miR-192-5p was analyzed by combining the differentially expressed mRNAs and the predicted genes and confirmed using a dual-luciferase reporter assay. It was found that miR-192-5p was found to regulate fat mass and obesity-associated (FTO) protein expression by directly targeting the 3' untranslated region of FTO mRNA. Moreover, in vivo and in vitro studies unveiled that FTO overexpression alleviated renal I/R injury and promoted HK-2 cell viability via stimulating autophagy flux. In conclusion, miR-192-5p aggravated I/R-induced renal injury by blocking autophagy flux via down-regulating FTO.
Collapse
Affiliation(s)
- Chengjun Zhang
- Center of Organ Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Ge Guan
- Center of Organ Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jiantao Wang
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Haijian Wei
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jinzhen Cai
- Center of Organ Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
5
|
Wu Y, Hu A, Shu X, Huang W, Zhang R, Xu Y, Yang C. Lactobacillus plantarum postbiotics trigger AMPK-dependent autophagy to suppress Salmonella intracellular infection and NLRP3 inflammasome activation. J Cell Physiol 2023; 238:1336-1353. [PMID: 37052047 DOI: 10.1002/jcp.31016] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
We previously found that Lactobacillus plantarum (LP)-derived postbiotics protected animals against Salmonella infection, but the molecular mechanism remains obscure. This study clarified the mechanisms from the perspective of autophagy. Intestinal porcine epithelial cells (IPEC-J2) were pretreated with LP-derived postbiotics (the culture supernatant, LPC; or heat-killed bacteria, LPB), and then challenged with Salmonella enterica Typhimurium (ST). Results showed that LP postbiotics markedly triggered autophagy under ST infection, as indicated by the increased LC3 and Beclin1 and the decreased p62 levels. Meanwhile, LP postbiotics (particularly LPC) exhibited a strong capacity of inhibiting ST adhesion, invasion and replication. Pretreatment with the autophagy inhibitor 3-methyladenine (3-MA) led to a significant decrease of autophagy and the aggravated infection, indicating the importance of autophagy in LP postbiotics-mediated Salmonella elimination. LP postbiotics (especially LPB) significantly suppressed ST-induced inflammation by modulating inflammatory cytokines (the increased interleukin (IL)-4 and IL-10, and decreased tumor necrosis factor-α (TNF), IL-1β, IL-6 and IL-18). Furthermore, LP postbiotics inhibited NOD-like receptor protein 3 (NLRP3) inflammasome activation, as evidenced by the decreased levels of NLRP3, Caspase-1 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). Deficits in autophagy resulted in an increase of inflammatory response and inflammasome activation. Finally, we found that both LPC and LPB triggered AMP-activated protein kinase (AMPK) signaling pathway to induce autophagy, and this was further confirmed by AMPK RNA interference. The intracellular infection and NLRP3 inflammasome were aggravated after AMPK knockdown. In summary, LP postbiotics trigger AMPK-mediated autophagy to suppress Salmonella intracellular infection and NLRP3 inflammasome in IPEC-J2 cells. Our findings highlight the effectiveness of postbiotics, and provide a new strategy for preventing Salmonella infection.
Collapse
Affiliation(s)
- Yanping Wu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Aixin Hu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Xin Shu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Wenxia Huang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Ruiqiang Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Yinglei Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Caimei Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| |
Collapse
|
6
|
Tiba AT, Qassam H, Hadi NR. Semaglutide in renal ischemia-reperfusion injury in mice. J Med Life 2023; 16:317-324. [PMID: 36937464 PMCID: PMC10015556 DOI: 10.25122/jml-2022-0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/13/2022] [Indexed: 03/21/2023] Open
Abstract
Ischemia and reperfusion injury (I/R) is a serious condition leading to organ failure, characterized by poor blood supply followed by rapid resuscitation of blood flow and reoxygenation. Renal failure caused by renal ischemia has high mortality and morbidity. This study aimed to explore the potential role of Semaglutide as a novel and effective therapeutic strategy for acute renal failure. Additionally, we aimed to assess the possible protective effect of Semaglutide on kidney I/R injury in mice through modulation of the inflammatory and oxidative pathways via phosphatidylinositol 3-kinase/adenosine triphosphate (PI3K/AKT) activation. We employed twenty-eight albino mice to induce the I/R injury model by clamping the renal artery for 30 min followed by a period of reperfusion for 2 hours. The control group was exposed to I/R injury, while the Semaglutide-treated group was pretreated with the drug 12 hours before induction of ischemia at a dose of 100 nmol/L/kg via the intraperitoneal route (i.p). In addition, the DMSO-treated group was subjected to similar conditions to the Semaglutide-treated group. At the end of the experiments, kidneys and blood samples were collected for investigation. Semaglutide could act as a protective agent against acute kidney injury by reducing inflammatory molecules such as tumor necrosis factor-alpha (TNF-α) and its cognate receptor, TNF-α R, interleukine-6 (IL-6). Furthermore, Semaglutide reduced F8 isoprostane levels, increased PI3K and AKT levels in renal tissues, and mitigated renal damage. Semaglutide had renoprotective effects via modulation of the inflammatory response and oxidative pathway by targeting the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Al-Tameemi Tiba
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Heider Qassam
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
- Corresponding Author: Najah Rayish Hadi, Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq. E-mail:
| |
Collapse
|
7
|
Zhang S, Xia W, Duan H, Li X, Qian S, Shen H. Ischemic Preconditioning Alleviates Mouse Renal Ischemia/Reperfusion Injury by Enhancing Autophagy Activity of Proximal Tubular Cells. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:217-230. [PMID: 35702707 PMCID: PMC9149508 DOI: 10.1159/000521850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/07/2022] [Indexed: 05/27/2023]
Abstract
OBJECTIVES Ischemia/reperfusion injury (IRI) is one of the most vital pathogenesis leading to kidney injury but lacks effective prevention and treatment strategies. This study was conducted to investigate the influences of ischemic preconditioning (IPC) on the pathological process of mouse renal IRI (RIRI) and to figure out the role of autophagy of proximal tubular cells (PTCs) in this process. METHODS C57BL/6J mice were randomized to three groups, i.e., sham-operated group, ischemia/reperfusion (I/R) group, and IPC + I/R group. Meanwhile, 3-methyladenine, an autophagy inhibitor, was administered when further verification was needed. Histological and functional severity of kidney injury, the autophagy and apoptosis activity of PTCs, as well as the characterization of the immune cell infiltration landscape in kidney tissues were investigated. Furthermore, HK-2 cells and primary cultured PTC were cultured to set up the hypoxic preconditioning and hypoxia/reoxygenation model for in vitro simulation and verification, and a microarray dataset derived from the Gene Expression Omnibus database was analyzed to explore the transcriptome profiles after IPC. RESULTS IPC could significantly attenuate I/R-induced kidney injury functionally and histologically both in the acute and recovery phase of RIRI by enhancing the autophagy activity of PTCs. Cell autophagy could regulate the release of monocyte chemoattractant protein-1, and sequentially decrease macrophages infiltration in kidney tissues in the acute phase of RIRI, thus mediating the reno-protective effect. CONCLUSIONS IPC could attenuate mouse RIRI-induced kidney injury. IPC-mediated activation of autophagy of PTCs plays a vital role in affording protection in RIRI-induced kidney injury.
Collapse
Affiliation(s)
- Shun Zhang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weimin Xia
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huangqi Duan
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyan Li
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Subo Qian
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haibo Shen
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Liu Y, Sun Y, Kang J, He Z, Liu Q, Wu J, Li D, Wang X, Tao Z, Guan X, She W, Xu H, Deng Y. Role of ROS-Induced NLRP3 Inflammasome Activation in the Formation of Calcium Oxalate Nephrolithiasis. Front Immunol 2022; 13:818625. [PMID: 35154136 PMCID: PMC8828488 DOI: 10.3389/fimmu.2022.818625] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/11/2022] [Indexed: 01/18/2023] Open
Abstract
Calcium oxalate nephrolithiasis is a common and highly recurrent disease in urology; however, its precise pathogenesis is still unknown. Recent research has shown that renal inflammatory injury as a result of the cell-crystal reaction plays a crucial role in the development of calcium oxalate kidney stones. An increasing amount of research have confirmed that inflammation mediated by the cell-crystal reaction can lead to inflammatory injury of renal cells, promote the intracellular expression of NADPH oxidase, induce extensive production of reactive oxygen species, activate NLRP3 inflammasome, discharge a great number of inflammatory factors, trigger inflammatory cascading reactions, promote the aggregation, nucleation and growth process of calcium salt crystals, and ultimately lead to the development of intrarenal crystals and even stones. The renal tubular epithelial cells (RTECs)-crystal reaction, macrophage-crystal reaction, calcifying nanoparticles, endoplasmic reticulum stress, autophagy activation, and other regulatory factors and mechanisms are involved in this process.
Collapse
Affiliation(s)
- Yunlong Liu
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan Sun
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Juening Kang
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziqi He
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Quan Liu
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jihua Wu
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Derong Li
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiang Wang
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhiwei Tao
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaofeng Guan
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wusheng She
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hua Xu
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaoliang Deng
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
9
|
Empagliflozin Enhances Autophagy, Mitochondrial Biogenesis, and Antioxidant Defense and Ameliorates Renal Ischemia/Reperfusion in Nondiabetic Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1197061. [PMID: 35126806 PMCID: PMC8816566 DOI: 10.1155/2022/1197061] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/16/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022]
Abstract
Background. Recent meta-analyses have shown that sodium-glucose cotransporter 2 (SGLT-2) inhibitors alleviate chronic kidney disease and acute kidney injury in diabetic patients. In this study, we aimed to investigate the effect of empagliflozin on renal ischemia/reperfusion (I/R) in nondiabetic rats and find the possible mechanisms. Experimental Approach. Eighteen male Wistar rats were randomly divided into three groups, including healthy control, ischemic control, and empagliflozin-treated group. Thirty minutes of bilateral renal ischemia was induced by clamping the renal hilum. Forty-eight hours after reopening the clamps, rats’ blood samples and tissue specimens were collected. Empagliflozin 10 mg/kg was administered by gavage, 2 hours before ischemia and 24 hours after the first dose. Results. I/R injury led to a significant rise in serum creatinine and blood urea nitrogen which was significantly decreased after treatment with empagliflozin. Empagliflozin also alleviated tubulointerstitial and glomerular damage and significantly decreased tissue histology scores. Empagliflozin decreased the increased levels of malondialdehyde, interleukin 1β, and tumor necrosis factor α. SGLT2 inhibition increased the decreased expression of nuclear factor erythroid 2-related factor 2 and PPARG coactivator 1 alpha that conduct antioxidant defense and mitochondrial biogenesis, respectively. Furthermore, empagliflozin markedly increased LC3-II/LC3-I and bcl2/bax ratios, showing its beneficial effect on activation of autophagy and inhibition of apoptosis. Despite its effects on diabetic nephropathy, empagliflozin did not activate the Sestrin2/AMP-activated protein kinase pathway in this study. Conclusion. Empagliflozin improved renal I/R injury in nondiabetic rats in this study by promoting autophagy and mitochondrial biogenesis and attenuation of oxidative stress, inflammation, and apoptosis.
Collapse
|
10
|
Li Y, Wang Z, Lian N, Wang Y, Zheng W, Xie K. Molecular Hydrogen: A Promising Adjunctive Strategy for the Treatment of the COVID-19. Front Med (Lausanne) 2021; 8:671215. [PMID: 34746162 PMCID: PMC8569706 DOI: 10.3389/fmed.2021.671215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an acute respiratory disease caused by a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has no specific and effective treatment. The pathophysiological process of the COVID-19 is an excessive inflammatory response after an organism infects with a virus. Inflammatory storms play an important role in the development of the COVID-19. A large number of studies have confirmed that hydrogen has a therapeutic effect on many diseases via inhibiting excessive inflammatory cells and factors. Recently, a study led by the Academician Zhong Nanshan in China on the treatment of the patients with the COVID-19 by inhalation of a mixed gas composed of hydrogen and oxygen has attracted widespread international attention and hydrogen therapy has also been included in a new treatment plan for the COVID-19 in China. This study mainly describes the mechanism of occurrence of the COVID-19, summarizes the therapeutic effects and underlying mechanisms of hydrogen on the critical disease, and analyzes the feasibility and potential therapeutic targets of hydrogen for the treatment of the COVID-19.
Collapse
Affiliation(s)
- Yingning Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Zhen Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Naqi Lian
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yuzun Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Weiqiang Zheng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China.,Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China.,College of Anesthesiology, Translational Research Institute of Intensive Care Medicine, College of Anesthesiology, Weifang Medical University, Weifang, China
| |
Collapse
|
11
|
PPARγ and TGFβ-Major Regulators of Metabolism, Inflammation, and Fibrosis in the Lungs and Kidneys. Int J Mol Sci 2021; 22:ijms221910431. [PMID: 34638771 PMCID: PMC8508998 DOI: 10.3390/ijms221910431] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a type II nuclear receptor, initially recognized in adipose tissue for its role in fatty acid storage and glucose metabolism. It promotes lipid uptake and adipogenesis by increasing insulin sensitivity and adiponectin release. Later, PPARγ was implicated in cardiac development and in critical conditions such as pulmonary arterial hypertension (PAH) and kidney failure. Recently, a cluster of different papers linked PPARγ signaling with another superfamily, the transforming growth factor beta (TGFβ), and its receptors, all of which play a major role in PAH and kidney failure. TGFβ is a multifunctional cytokine that drives inflammation, fibrosis, and cell differentiation while PPARγ activation reverses these adverse events in many models. Such opposite biological effects emphasize the delicate balance and complex crosstalk between PPARγ and TGFβ. Based on solid experimental and clinical evidence, the present review summarizes connections and their implications for PAH and kidney failure, highlighting the similarities and differences between lung and kidney mechanisms as well as discussing the therapeutic potential of PPARγ agonist pioglitazone.
Collapse
|
12
|
Qi B, Yu Y, Wang Y, Wang Y, Yu Y, Xie K. Perspective of Molecular Hydrogen in the Treatment of Sepsis. Curr Pharm Des 2021; 27:667-678. [PMID: 32912119 DOI: 10.2174/1381612826666200909124936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/09/2020] [Indexed: 11/22/2022]
Abstract
Sepsis is the main cause of death in critically ill patients with no effective treatment. Sepsis is lifethreatening organ dysfunction due to a dysregulated host response to infection. As a novel medical gas, molecular hydrogen (H2) has a therapeutic effect on many diseases, such as sepsis. H2 treatment exerts multiple biological effects, which can effectively improve multiple organ injuries caused by sepsis. However, the underlying molecular mechanisms of hydrogen involved in the treatment of sepsis remain elusive, which are likely related to anti-inflammation, anti-oxidation, anti-apoptosis, regulation of autophagy and multiple signaling pathways. This review can help better understand the progress of hydrogen in the treatment of sepsis, and provide a theoretical basis for the clinical application of hydrogen therapy in sepsis in the future.
Collapse
Affiliation(s)
- Bo Qi
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yaoqi Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuzun Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
13
|
Vázquez-Carballo C, Guerrero-Hue M, García-Caballero C, Rayego-Mateos S, Opazo-Ríos L, Morgado-Pascual JL, Herencia-Bellido C, Vallejo-Mudarra M, Cortegano I, Gaspar ML, de Andrés B, Egido J, Moreno JA. Toll-Like Receptors in Acute Kidney Injury. Int J Mol Sci 2021; 22:ijms22020816. [PMID: 33467524 PMCID: PMC7830297 DOI: 10.3390/ijms22020816] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is an important health problem, affecting 13.3 million individuals/year. It is associated with increased mortality, mainly in low- and middle-income countries, where renal replacement therapy is limited. Moreover, survivors show adverse long-term outcomes, including increased risk of developing recurrent AKI bouts, cardiovascular events, and chronic kidney disease. However, there are no specific treatments to decrease the adverse consequences of AKI. Epidemiological and preclinical studies show the pathological role of inflammation in AKI, not only at the acute phase but also in the progression to chronic kidney disease. Toll-like receptors (TLRs) are key regulators of the inflammatory response and have been associated to many cellular processes activated during AKI. For that reason, a number of anti-inflammatory agents targeting TLRs have been analyzed in preclinical studies to decrease renal damage during AKI. In this review, we updated recent knowledge about the role of TLRs, mainly TLR4, in the initiation and development of AKI as well as novel compounds targeting these molecules to diminish kidney injury associated to this pathological condition.
Collapse
Affiliation(s)
- Cristina Vázquez-Carballo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
| | - Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Sandra Rayego-Mateos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
| | - Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - José Luis Morgado-Pascual
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Carmen Herencia-Bellido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
| | - Mercedes Vallejo-Mudarra
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Isabel Cortegano
- Immunobiology Department, Carlos III Health Institute, 28220 Majadahonda (Madrid), Spain; (I.C.); (M.L.G.); (B.d.A.)
| | - María Luisa Gaspar
- Immunobiology Department, Carlos III Health Institute, 28220 Majadahonda (Madrid), Spain; (I.C.); (M.L.G.); (B.d.A.)
| | - Belén de Andrés
- Immunobiology Department, Carlos III Health Institute, 28220 Majadahonda (Madrid), Spain; (I.C.); (M.L.G.); (B.d.A.)
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
- Correspondence: (J.E.); (J.A.M.); Tel.: +34-915504800 (J.E.); +34-957-218039 (J.A.M.)
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 28029 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 140471 Cordoba, Spain
- Correspondence: (J.E.); (J.A.M.); Tel.: +34-915504800 (J.E.); +34-957-218039 (J.A.M.)
| |
Collapse
|
14
|
Li S, Shi M, Wan Y, Wang Y, Zhu M, Wang B, Zhan Y, Ran B, Wu C. Inflammasome/NF-κB translocation inhibition via PPARγ agonist mitigates inorganic mercury induced nephrotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 201:110801. [PMID: 32502906 DOI: 10.1016/j.ecoenv.2020.110801] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/30/2020] [Accepted: 05/24/2020] [Indexed: 06/11/2023]
Abstract
Mercury (Hg) pollution poses global human health and environmental risks. However, still knowledge gaps exist on both exposures and health effects. Here, we combined transcriptome sequencing technique to further investigate the specific mechanisms of inorganic Hg toxicity in the kidney. Strikingly, transcriptomic analysis revealed that 4174 unigenes (including 2646 upregulated and 1528 downregulated unigenes) were differentially expressed under acute HgCl2 (5 mg/kg) exposure in the kidney. Additionally, we observed that HgCl2 selectively induced tumor necrosis factor superfamily (TNFSF) to participate in renal damage, which was consistent with the high-throughput sequencing data. The phenomenon is accompanied by NLRP3 inflammasome and NF-κB signal activation in the kidney. Simultaneously, ELISA results shown that TNF-α, IL-1β and IL-6 concentrations in the kidney were significant increased. KEGG enrichment analysis showed that peroxisome proliferators-activated receptors (PPAR) signaling pathway might be vital toxic mechanism of Hg in the kidney. Then, our data showed that PPARγ agonist (GW 1929) attenuated HgCl2 (15 μg/ml)-induced apoptosis and NLRP3 inflammasome activation via decreasing translocation of NF-κB and increasing Bcl2 levels in vitro. Along with this, we demonstrated that PPARγ antagonists (GW9662) effectively aggravated HgCl2-induced nephrotoxicity. Overall, our results suggested that PPARγ signaling pathway is considered to be a protective mechanism to combat against HgCl2-triggered NLRP3 inflammasome activation and apoptosis.
Collapse
Affiliation(s)
- Siwen Li
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Mei Shi
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Ying Wan
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Yanling Wang
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Mei Zhu
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Boya Wang
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Yangmei Zhan
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Bing Ran
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Chunling Wu
- College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China.
| |
Collapse
|
15
|
AvrA Exerts Inhibition of NF-κB Pathway in Its Naïve Salmonella Serotype through Suppression of p-JNK and Beclin-1 Molecules. Int J Mol Sci 2020; 21:ijms21176063. [PMID: 32842467 PMCID: PMC7504150 DOI: 10.3390/ijms21176063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 01/02/2023] Open
Abstract
Avian salmonellosis caused by Salmonella enterica serovar Enteritidis (S. Enteritidis) and Pullorum (S. Pullorum) remains a big threat to the poultry industry and public hygiene. AvrA is an effector involved in inhibiting inflammation. Compared to AvrA from S. Enteritidis (SE-AvrA), the AvrA from S. Pullorum (SP-AvrA) lacks ten amino acids at the C-terminal. In this study, we compared the anti-inflammatory response induced by SP-AvrA to that of SE-AvrA. Transient expression of SP-AvrA in epithelial cells resulted in significantly weaker inhibition of NF-κB pathway activation when treated with TNF-α compared to the inhibition by SE-AvrA. SP-AvrA expression in the S. Enteritidis resulted in weaker suppression of NF-κB pathway in infected HeLa cells compared to SE-AvrA expression in the cells, while SP-AvrA expressed in S. Pullorum C79-13 suppressed NF-κB activation in infected HeLa and Caco 2 BBE cells to a greater extent than did SE-AvrA because of the higher expression of SP-AvrA than SE-AvrA in S. Pullorum. Further analysis demonstrated that the inhibition of NF-κB pathway in Salmonella-infected cells corresponded to the downregulation of the p-JNK and Beclin-1 protein molecules. Our study reveals that AvrA modifies the anti-inflammatory response in a manner dependent on the Salmonella serotype through inhibition of NF-κB pathway.
Collapse
|
16
|
Dapagliflozin improves behavioral dysfunction of Huntington's disease in rats via inhibiting apoptosis-related glycolysis. Life Sci 2020; 257:118076. [PMID: 32659371 DOI: 10.1016/j.lfs.2020.118076] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/24/2020] [Accepted: 07/07/2020] [Indexed: 12/31/2022]
Abstract
AIMS Huntington's disease is a rare neurodegenerative disorder which is associated with defected glucose metabolism with consequent behavioral disturbance including memory and locomotion. 3-nitropropionic acid (3-NP) can cause, in high single dose, an acute striatal injury/Huntington's disease. Dapagliflozin, which is one of the longest duration of action of SGLTIs family, may be able to diminish that injury and its resultant behavioral disturbances. MATERIAL AND METHODS Forty rats were divided into four groups (n = 10 in each group): normal control group (CTRL), dapagliflozin (CTRL + DAPA) group, 3-nitropropionic acid (3-NP) group, and dapagliflozin plus 3-nitropropionic acid (DAPA + 3-NP) group. Behavioral tests (beam walking test, hanging wire test, limb withdrawal test, Y-maze spontaneous alteration, elevated plus maze) were performed with evaluating neurological scoring. In striatum, neurotransmitters (glutamate, aspartate, GABA, ACh and AChE activity) were measured. In addition, apoptosis and glycolysis markers (NF-κB, Cyt-c, lactate, HK-II activity, P53, calpain, PEA15 and TIGAR) were determined. Inflammation (IL-1β, IL-6, IL-8 and TNF-α) and autophagy (beclin-1, LC3 and DRAM) indicators were measured. Additionally, histopathological screening was conducted. KEY FINDINGS 3-Nitropropionic acid had the ability to perturb the neurotransmission which was reflected in impaired behavioral outcome. All of glycolysis, apoptosis and inflammation markers were elevated after 3-NP acute intoxication but autophagy parameters, except DRAM, were reduced. However, DAPA markedly reversed the abovementioned parameters. SIGNIFICANCE Dapagliflozin demonstrated anti-glycolytic, anti-apoptotic, anti-inflammatory and autophagic effects on 3-NP-damaged striatal cells and promoted the behavioral outcome.
Collapse
|
17
|
Enhancing Autophagy Protects Against Sepsis-Induced Neuromuscular Dysfunction Associated with Qualitative Changes to Acetylcholine Receptors. Shock 2020; 52:111-121. [PMID: 30286033 DOI: 10.1097/shk.0000000000001189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Sepsis-induced myopathy is a heavy burden for patients during respiratory failure as well as after discharge, which could be characterized with qualitative changes to nAChR in a rat model of sepsis, regulated by NRG-1. Autophagy is an innate immune defense mechanism against microbial challenges. We found neuromuscular dysfunction in anterior tibial muscle of male Sprague-Dawley rats 24 h after cecal ligation and puncture (CLP). CLP resulted in increased systemic and local inflammation in anterior tibial muscle tissue. The start-up phase of autophagy, as measured by LC3II, was activated immediately after CLP and continued until 24 h; the degradation phase was suppressed until 24 h, after a brief increase at 4 h (revealed by p62). NRG-1 first increased, and then decreased to a level lower than that in the sham group. Meanwhile, expression of γ- and α7- acetylcholine receptors was detected at 8 and 16 h after CLP; levels increased continuously until 24 h. Subsequently, we investigated the significance of autophagy in CLP-induced neuromuscular dysfunction by treatment with rapamycin or 3-methyladenine, which were classical pharmaceuticals for enhancing or suppressing autophagy. Rapamycin activated autophagy, limited the CLP-induced systemic pro-inflammatory response and blood bacterial load without affecting local inflammatory response, upregulated NRG-1, downregulated γ- and α7-acetylcholine receptors, and improved 7-day neuromuscular function and survival rate. In contrast, 3-methyladenine enhanced local inflammatory response, suppressed autophagy, worsened 7-day neuromuscular function. We conclude that impaired autophagy may contribute to sepsis-induced neuromuscular dysfunction in young male rats. Enhancing autophagy with rapamycin alleviated qualitative changes to acetylcholine receptors without triggering local anti-inflammatory response and improved anterior tibial muscle function in septic early phase (24 h) as well as in septic chronic phase (7d). Enhancing autophagy soon after sepsis is a potential strategy for treatment of sepsis-induced myopathy.
Collapse
|
18
|
Li C, Li C, Lin J, Zhao G, Xu Q, Jiang N, Wang Q, Peng X, Zhu G, Jiang J. The Role of Autophagy in the Innate Immune Response to Fungal Keratitis Caused by Aspergillus fumigatus Infection. Invest Ophthalmol Vis Sci 2020; 61:25. [PMID: 32084267 PMCID: PMC7326573 DOI: 10.1167/iovs.61.2.25] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose To determine the role of autophagy in the innate immune response to fungal keratitis (FK) caused by Aspergillus fumigatus infection. Methods Corneal samples obtained from patients and mice with FK were visualized via transmission electron microscopy (TEM). Autophagy-related proteins LC3B-II, Beclin-1, LAMP-1, and p62 in A. fumigatus-infected corneas of C57BL/6 mice were tested by Western blot. After treatment with autophagy inhibitors 3-methyladenine (3-MA), chloroquine (CQ), or inducer rapamycin, autophagy-related proteins were detected by Western blot. Corneas were photographed with slit lamp microscopy and pathological changes were observed by hematoxylin and eosin staining. Polymorphonuclear neutrophilic leukocytes (PMNs) were assessed by immunofluorescent staining and observed under TEM. The levels of CXCL-1, IL-1β, HMGB1, IL-18, TNF-α, and IL-10 were tested by reverse transcription polymerase chain reaction and Western blot. The quantification of fungal loads was detected and photographed. Results The accumulation of autophagosomes in corneas of patients and mice with FK was observed with TEM. The expression of LC3B-II, Beclin-1, and LAMP-1 was elevated in corneas after fungal infection, whereas p62 was reduced. Treatment with 3-MA or CQ upregulated clinical scores, pathological changes, and the expression of CXCL-1, IL-1β, HMGB1, IL-18, and TNF-α except IL-10. The morphology of PMNs was changed and PMN recruitment was increased in mice corneas treated with 3-MA or CQ, whereas rapamycin reduced the inflammatory response to keratitis. These results were statistically significant. Conclusions A. fumigatus infection increases the expression of autophagy in corneas. Autophagy plays an anti-inflammatory role in the innate immune response to A. fumigatus keratitis.
Collapse
|
19
|
Li XY, Chen HR, Zha XQ, Chen S, Pan LH, Li QM, Luo JP. Prevention and possible mechanism of a purified Laminaria japonica polysaccharide on adriamycin-induced acute kidney injury in mice. Int J Biol Macromol 2020; 148:591-600. [PMID: 31958563 DOI: 10.1016/j.ijbiomac.2020.01.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/26/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022]
Abstract
The present work aims to investigate the effects and underlying mechanism of a homogeneous Laminaria japonica polysaccharide (LJP61A) on acute kidney injury (AKI) in mice. According to the results of biochemical and pathological analysis, we concluded that LJP61A could protect kidney from the damage of adriamycin in AKI mice. Compared to the model group, the mRNA level of cytokines (TNF-α, IL-1β and MCP-1) and protein level of mesenchymal markers demsin were decrease by the treatment of LJP61A while the protein levels of podocyte structure markers (Nephrin and WT-1) were increased. Moreover, the adriamycin-induced enhancement of phosphor-p65, phosphor-p38, phosphor-ERK1/2 and phosphor-JNK in the kidney of AKI mice were significantly suppressed by LJP61A. Similar variation was observed in the mRNA and protein levels of TGF-β1 and Smad3. These results suggested that LJP61A prevented acute kidney injury possibly via regulating TGF-β1-mediated Smad3, MAPKs and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Hao-Ran Chen
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China; Key Laboratory of Metabolism and Regulation for Major Disease of Anhui Higher Education Institutes, Hefei University of Technology, People's Republic of China.
| | - Shun Chen
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Li-Hua Pan
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China.
| |
Collapse
|
20
|
Characteristics of Changes in Inflammatory Cytokines as a Function of Hepatic Ischemia-Reperfusion Injury Stage in Mice. Inflammation 2020; 42:2139-2147. [PMID: 31494794 DOI: 10.1007/s10753-019-01078-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver ischemia-reperfusion injury (IRI) can severely compromise the prognosis of patients receiving liver surgery. While inflammation contributes to the damage resulting from IRI, only a limited number of inflammation biomarkers have been identified as being associated with the different stages of hepatic IRI. As an approach to identify some of these inflammatory cytokines and the molecular mechanisms involved within different stages of hepatic IRI, we used an advanced antibody array assay to detect multiple proteins. With this technology, we observed specific differences in the content of inflammatory cytokines between ischemic and sham controls, as well as a function of the different reperfusion stages in a hepatic IRI mouse model. For example, while liver tissue content of IL-12p40/p70 was significantly increased in the ischemic stage, it was significantly decreased in the reperfusion stage as compared with that of the sham group. For other inflammatory cytokines, no changes were obtained between the ischemic and reperfusion stages with levels of IL-17, Eotaxin-2, Eotaxin, and sTNF RII all being consistently increased, while those of TIMP-1, TIMP-2, BLC, and MCSF consistently decreased as compared with that of the sham group at all reperfusion stages examined. Results from protein function annotation Gene Ontology and the KEGG pathway revealed that inflammatory cytokines are enriched in a network associated with activation of inflammatory response signaling pathways such as TLR, TNF, and IL-17 when comparing responses of the IR versus sham groups. The identification of cytokines along with their roles at specific stages of IRI may reveal important new biological markers for the diagnosis and prognosis of hepatic IRI.
Collapse
|
21
|
Kang Y, Li Y, Wen H, Zhu J, Zheng J, Feng Z. Prevention of renal ischemia and reperfusion injury by penehyclidine hydrochloride through autophagy activation. Mol Med Rep 2020; 21:2182-2192. [PMID: 32186764 PMCID: PMC7115187 DOI: 10.3892/mmr.2020.11024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/29/2019] [Indexed: 11/06/2022] Open
Abstract
Penehyclidine hydrochloride (PHC) suppresses renal ischemia and reperfusion (I/R) injury (IRI); however, the underlying mechanism of action that achieves this function remains largely unknown. The present study aimed to investigate the potential role of autophagy in PHC‑induced suppression of renal IRI, as well as the involvement of cell proliferation and apoptosis. A rat IRI model and a cellular hypoxia/oxygenation (H/R) model were established; PHC, 3‑methyladenine (3‑MA) and rapamycin (Rapa) were administered to the IRI model rats prior to I/R induction and to H/R cells following reperfusion. Serum creatinine was measured using a biochemistry analyzer, whereas aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT) expression levels were detected using ELISA kits. Renal tissue injury was evaluated by histological examination. In addition, microtubule‑associated protein light chain 3B (LC3B) expression, autophagosome formation, cell proliferation and apoptosis were detected in the cellular H/R model. The results demonstrated that I/R induced renal injury in IRI model rats, upregulated serum creatinine, ALAT and ASAT expression levels, and increased autophagic processes. In contrast, pretreatment with PHC or Rapa significantly prevented these I/R‑induced changes, whereas the administration of 3‑MA enhanced I/R‑induced injuries through suppressing autophagy. PHC and Rapa increased LC3B and Beclin‑1 expression levels, but decreased sequestome 1 (p62) expression in the cellular H/R model, whereas 3‑MA prevented these PHC‑induced changes. PHC and Rapa promoted proliferation and autophagy in the cellular H/R model; these effects were accompanied by increased expression levels of LC3B and Beclin‑1, and reduced p62 expression levels, whereas these levels were inhibited by 3‑MA. Furthermore, PHC and Rapa inhibited apoptosis in the cellular H/R model through increasing Bcl‑2 expression levels, and suppressing Bax and caspase‑3 expression levels; the opposite effect was induced by 3‑MA. In conclusion, PHC suppressed renal IRI through the induction of autophagy, which in turn promoted proliferation and suppressed apoptosis in renal cells.
Collapse
Affiliation(s)
- Yuqing Kang
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai 201599, P.R. China
| | - Yuebing Li
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310005, P.R. China
| | - Heng Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310005, P.R. China
| | - Junfeng Zhu
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai 201599, P.R. China
| | - Jiangbo Zheng
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai 201599, P.R. China
| | - Zhaoming Feng
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai 201599, P.R. China
| |
Collapse
|
22
|
Sefi M, Chaâbane M, Elwej A, Bejaoui S, Marrekchi R, Jamoussi K, Gouiaa N, Boudawara Sellami T, El Cafsi M, Zeghal N, Soudani N. Zinc alleviates maneb-induced kidney injury in adult mice through modulation of oxidative stress, genotoxicity, and histopathological changes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:8091-8102. [PMID: 31897980 DOI: 10.1007/s11356-019-07175-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/25/2019] [Indexed: 06/10/2023]
Abstract
Zinc is one of the important essential trace minerals to human health due to its antioxidant properties. The present study was conducted to elucidate its potential protective role against maneb-induced nephrotoxicity. For this purpose, animals were randomly divided into four groups of six each. Mice of group I (negative controls) have received daily 0.5 ml of distilled water, a solvent of maneb. Mice of group II (MB) have received 30 mg/kg bw of maneb daily by intraperitoneal way. Mice of group III (MB + Zn) have received the same dose of maneb as group II, along with ZnSO4 (30 mg/kg bw) daily. Mice of group IV (Zn), considered as positive controls, have received the same dose of ZnSO4 as group III daily. Our results revealed that ZnSO4 co-administration to maneb-treated mice decreased kidney levels of malondialdehyde, hydrogen peroxide, protein carbonyls, and advanced oxidation protein products; the levels of non-enzymatic antioxidants like vitamin C, glutathione, and metallothionein. It recovered the alteration of antioxidant enzyme activities (catalase, superoxide dismutase, and glutathione peroxidase) and attenuated DNA fragmentation. Furthermore, this essential trace element was also able to alleviate kidney biomarkers' alterations by lowering plasma levels of creatinine, urea, uric acid, and lactate dehydrogenase. In addition, the histopathological changes induced by maneb were improved following zinc administration. Our results indicated that zinc might be beneficial against maneb-induced renal oxidative damage in mice.
Collapse
Affiliation(s)
- Mediha Sefi
- Animal Physiology Laboratory, Department of Life Sciences, University of Sfax, Sciences Faculty of Sfax, Sfax, Tunisia.
- Ecology, Biology and physiology laboratory of aquatic organisms, Department of Biological Sciences, Sciences Faculty of Tunis, University Tunis El Manar, 2092, Tunis, Tunisia.
| | - Mariem Chaâbane
- Animal Physiology Laboratory, Department of Life Sciences, University of Sfax, Sciences Faculty of Sfax, Sfax, Tunisia
| | - Awatef Elwej
- Animal Physiology Laboratory, Department of Life Sciences, University of Sfax, Sciences Faculty of Sfax, Sfax, Tunisia
| | - Safa Bejaoui
- Ecology, Biology and physiology laboratory of aquatic organisms, Department of Biological Sciences, Sciences Faculty of Tunis, University Tunis El Manar, 2092, Tunis, Tunisia
| | - Rim Marrekchi
- Biochemistry Laboratory, Department of Biochemistry, CHU Hedi Chaker, University of Sfax, 3029, Sfax, Tunisia
| | - Kamel Jamoussi
- Biochemistry Laboratory, Department of Biochemistry, CHU Hedi Chaker, University of Sfax, 3029, Sfax, Tunisia
| | - Naourez Gouiaa
- Histopathology Laboratory, Department of Anatomo-pathology, CHU Habib Bourguiba, University of Sfax, Sfax, Tunisia
| | - Tahia Boudawara Sellami
- Histopathology Laboratory, Department of Anatomo-pathology, CHU Habib Bourguiba, University of Sfax, Sfax, Tunisia
| | - M'hamed El Cafsi
- Ecology, Biology and physiology laboratory of aquatic organisms, Department of Biological Sciences, Sciences Faculty of Tunis, University Tunis El Manar, 2092, Tunis, Tunisia
| | - Najiba Zeghal
- Animal Physiology Laboratory, Department of Life Sciences, University of Sfax, Sciences Faculty of Sfax, Sfax, Tunisia
| | - Nejla Soudani
- Animal Physiology Laboratory, Department of Life Sciences, University of Sfax, Sciences Faculty of Sfax, Sfax, Tunisia
- Ecology, Biology and physiology laboratory of aquatic organisms, Department of Biological Sciences, Sciences Faculty of Tunis, University Tunis El Manar, 2092, Tunis, Tunisia
| |
Collapse
|
23
|
Activation of sirtuin1 protects against ischemia/reperfusion-induced acute kidney injury. Biomed Pharmacother 2020; 125:110021. [PMID: 32092826 DOI: 10.1016/j.biopha.2020.110021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/07/2020] [Accepted: 02/13/2020] [Indexed: 12/16/2022] Open
Abstract
Sirtuin1 (SIRT1), a class III histone deacetylase, exerts a protective role against kidney injury. However, its functions in renal ischemia/reperfusion (I/R) injury remains unclear as yet. In this study, we established acute kidney injury (AKI) rat model through renal ischemia and reperfusion, and the role of SIRT1 in I/R-induced AKI was investigated both in vivo and in vitro. In in vivo study, SIRT1 was expressed in tubular epithelial cells (TECs) and its expression was upregulated after I/R treatment. Meanwhile, our in vitro experiment confirmed that the expression of SIRT1 was also elevated in human renal proximal tubular epithelial (HK2) cells treated with hypoxia and reoxygenation (H/R). Notably, activation of SIRT1 by resveratrol (Res, an activator of SIRT1) could significantly ameliorate renal function and reduce the TECs apoptosis in rats. Likewise, Res intervention also reduced the apoptosis and the production of reactive oxygen species in HK2 cells. Furthermore, we found that the autophagy level was upregulated in I/R injury, which could be raised further through resveratrol intervention; and chloroquine (CQ, an autophagy inhibitor) did reverse these protective effects of SIRT1 activation. Taken together, our results suggest that SIRT1 plays a protective role by autophagy induction in I/R- induced AKI. Its role might serve as a preventive approach in I/R-associated AKI.
Collapse
|
24
|
Liu S, Yang Y, Gao H, Zhou N, Wang P, Zhang Y, Zhang A, Jia Z, Huang S. Trehalose attenuates renal ischemia-reperfusion injury by enhancing autophagy and inhibiting oxidative stress and inflammation. Am J Physiol Renal Physiol 2020; 318:F994-F1005. [PMID: 32068461 DOI: 10.1152/ajprenal.00568.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Renal ischemia-reperfusion (IR) injury is one of the most common acute kidney injuries, but there is still a lack of effective treatment in the clinical setting. Trehalose (Tre), a natural disaccharide, has been demonstrated to protect against oxidative stress, inflammation, and apoptosis. However, whether it could protect against IR-induced renal injury needs to be investigated. In an in vivo experiment, C57BL/6J mice were pretreated with or without Tre (2 g/kg) through a daily single intraperitoneal injection from 3 days before renal IR surgery. Renal function, apoptosis, oxidative stress, and inflammation were analyzed to evaluate kidney injury. In an in vitro experiment, mouse proximal tubular cells were treated with or without Tre under a hypoxia/reoxygenation condition. Western blot analysis, autophagy flux detection, and apoptosis assay were performed to evaluate the level of autophagy and antiapoptotic effect of Tre. The in vivo results showed that the renal damage induced by IR was ameliorated by Tre treatment, as renal histology and renal function were improved and the enhanced protein levels of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin were blocked. Moreover, autophagy was activated by Tre pretreatment along with inhibition of the IR injury-induced apoptosis, oxidative stress, and inflammation. The in vitro results showed that Tre treatment activated autophagy and protected against hypoxia/reoxygenation-induced tubular cell apoptosis and oxidative stress. Our results demonstrated that Tre protects against IR-induced renal injury, possibly by enhancing autophagy and blocking oxidative stress, inflammation, and apoptosis, suggesting its potential use for the clinical treatment of renal IR injury.
Collapse
Affiliation(s)
- Suwen Liu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yunwen Yang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Huiping Gao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Ning Zhou
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Peipei Wang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Andrianova NV, Zorova LD, Babenko VA, Pevzner IB, Popkov VA, Silachev DN, Plotnikov EY, Zorov DB. Rapamycin Is Not Protective against Ischemic and Cisplatin-Induced Kidney Injury. BIOCHEMISTRY (MOSCOW) 2020; 84:1502-1512. [PMID: 31870254 DOI: 10.1134/s0006297919120095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autophagy plays an important role in the pathogenesis of acute kidney injury (AKI). Although autophagy activation was shown to be associated with an increased lifespan and beneficial effects in various pathologies, the impact of autophagy activators, particularly, rapamycin and its analogues on AKI remains obscure. In our study, we explored the effects of rapamycin treatment in in vivo and in vitro models of ischemic and cisplatin-induced AKI. The impact of rapamycin on the kidney function after renal ischemia/reperfusion (I/R) or exposure to the nephrotoxic agent cisplatin was assessed by quantifying blood urea nitrogen and serum creatinine and evaluating the content of neutrophil gelatinase-associated lipocalin, a novel biomarker of AKI. In vitro experiments were performed on the primary culture of renal tubular cells (RTCs) that were subjected to oxygen-glucose deprivation (OGD) or incubated with cisplatin under various rapamycin treatment protocols. Cell viability and proliferation were estimated by the MTT assay and real-time cell analysis using an RTCA iCELLigence system. Although rapamycin inhibited mTOR (mammalian target of rapamycin) signaling, it failed to enhance the autophagy and to ameliorate the severity of AKI caused by ischemia or cisplatin-induced nephrotoxicity. Experiments with RTCs demonstrated that rapamycin exhibited the anti-proliferative effect in primary RTCs cultures but did not protect renal cells exposed to OGD or cisplatin. Our study revealed for the first time that the mTOR inhibitor rapamycin did not prevent AKI caused by renal I/R or cisplatin-induced nephrotoxicity and, therefore, cannot be considered as an ideal mimetic of the autophagy-associated nephroprotective mechanisms (e.g., those induced by caloric restriction), as it had been suggested earlier. The protective action of such approaches like caloric restriction might not be limited to mTOR inhibition and can proceed through more complex mechanisms involving alternative autophagy-related targets. Thus, the use of rapamycin and its analogues for the treatment of various AKI forms requires further studies in order to understand potential protective or adverse effects of these compounds in different contexts.
Collapse
Affiliation(s)
- N V Andrianova
- Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, 119992, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - L D Zorova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - V A Babenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - I B Pevzner
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - V A Popkov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - D N Silachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| | - E Y Plotnikov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia. .,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia.,Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Moscow, 119991, Russia
| | - D B Zorov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia. .,Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow, 117997, Russia
| |
Collapse
|
26
|
Bhatia D, Choi ME. Autophagy in kidney disease: Advances and therapeutic potential. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:107-133. [PMID: 32620239 DOI: 10.1016/bs.pmbts.2020.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is a highly conserved intracellular catabolic process for the degradation of cytoplasmic components that has recently gained increasing attention for its importance in kidney diseases. It is indispensable for the maintenance of kidney homeostasis both in physiological and pathological conditions. Investigations utilizing various kidney cell-specific conditional autophagy-related gene knockouts have facilitated the advancement in understanding of the role of autophagy in the kidney. Recent findings are raising the possibility that defective autophagy exerts a critical role in different pathological conditions of the kidney. An emerging body of evidence reveals that autophagy exhibits cytoprotective functions in both glomerular and tubular compartments of the kidney, suggesting the upregulation of autophagy as an attractive therapeutic strategy. However, there is also accumulating evidence that autophagy could be deleterious, which presents a formidable challenge in developing therapeutic strategies targeting autophagy. Here, we review the recent advances in research on the role of autophagy during different pathological conditions, including acute kidney injury (AKI), focusing on sepsis, ischemia-reperfusion injury, cisplatin, and heavy metal-induced AKI. We also discuss the role of autophagy in chronic kidney disease (CKD) focusing on the pathogenesis of tubulointerstitial fibrosis, podocytopathies including focal segmental glomerulosclerosis, diabetic nephropathy, IgA nephropathy, membranous nephropathy, HIV-associated nephropathy, and polycystic kidney disease.
Collapse
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
27
|
Jin J, Gong J, Zhao L, Zhang H, He Q, Jiang X. Inhibition of high mobility group box 1 (HMGB1) attenuates podocyte apoptosis and epithelial-mesenchymal transition by regulating autophagy flux. J Diabetes 2019; 11:826-836. [PMID: 30864227 DOI: 10.1111/1753-0407.12914] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 02/11/2019] [Accepted: 02/27/2019] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Podocyte injury, characterized by podocyte hypertrophy, apoptosis, and epithelial-mesenchymal transition (EMT), is the major causative factor of diabetic nephropathy (DN). Autophagy dysfunction is regarded as the major risk factor for podocyte injury including EMT and apoptosis. High mobility group box 1 (HMGB1) is involved in the progression of DN through the induction of autophagy. However, the underlying mechanism remains unknown. METHODS Plasma HMGB1 concentrations were determined in DN patients using ELISA. Apoptosis of DN serum-treated podocytes was evaluated by flow cytometry. Podocyte autophagy flux was measured using immunofluorescence. Western blotting analysis was used to investigate HMGB1 expression, EMT, and autophagy-related signaling pathways. RESULTS Upregulation of HMGB1 was found in DN patients and DN serum-treated podocytes. Removal of HMGB1 inhibited DN serum-mediated podocyte apoptosis by inhibiting autophagy and activating AKT/mammalian target of rapamycin (mTOR) signaling. In addition, HMGB1 depletion repressed the progression of podocyte EMT by inhibiting transforming growth factor (TGF)-β/smad1 signaling in vitro and in vivo. The combination of HMGB1 short interference (si) RNA and the autophagy activator rapamycin protected against podocyte apoptosis and EMT progression by inhibiting the AKT/mTOR and TGF-β/smad signaling pathway, respectively. CONCLUSIONS Although HMGB1 siRNA and rapamycin treatment had opposite effects on autophagy and AKT/mTOR signaling, there was no contradiction about the role of HMGB1 siRNA and rapamycin on AKT/mTOR pathway because autophagy and AKT/mTOR signaling play dual roles in intracellular biological processes. Based on the findings of this study, we may assume that HMGB1-initiated autophagy is harmful, whereas rapamycin is beneficial to podocyte survival. Possibly combined treatment with HMGB1 siRNA and rapamycin improved podocyte damage and EMT by regulating autophagy homeostasis.
Collapse
Affiliation(s)
- Juan Jin
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, China
- Department of Nephrology, People's Hospital of Hangzhou Medical College, Zhejiang, China
| | - Jianguang Gong
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, China
- Department of Nephrology, People's Hospital of Hangzhou Medical College, Zhejiang, China
| | - Li Zhao
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, China
- Department of Nephrology, People's Hospital of Hangzhou Medical College, Zhejiang, China
| | - Hongjuan Zhang
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, China
- Department of Nephrology, People's Hospital of Hangzhou Medical College, Zhejiang, China
| | - Qiang He
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, China
- Department of Nephrology, People's Hospital of Hangzhou Medical College, Zhejiang, China
| | - Xinxin Jiang
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, China
- Department of Nephrology, People's Hospital of Hangzhou Medical College, Zhejiang, China
| |
Collapse
|
28
|
Xi X, Zou C, Ye Z, Huang Y, Chen T, Hu H. Pioglitazone protects tubular cells against hypoxia/reoxygenation injury through enhancing autophagy via AMPK-mTOR signaling pathway. Eur J Pharmacol 2019; 863:172695. [PMID: 31560869 DOI: 10.1016/j.ejphar.2019.172695] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/25/2022]
Abstract
Pioglitazone (Pio), a peroxisome proliferators-activated receptor-γ (PPAR-γ) agonist, may protect against renal ischemia/reperfusion injury (IRI). Recent studies have shown that autophagy plays a protective role in IRI. We aimed to evaluate whether autophagy was involved in pioglitazone-induced protection during tubular cell hypoxia/reoxygenation (H/R). Normal rat kidney proximal tubular cells NRK-52E were subjected to H/R injury, and they were divided into 6 groups: control, control + Pio, H/R, H/R + Pio, H/R + MA, H/R + MA + Pio. Autophagy-related proteins were primarily assessed by Western blot and TUNEL was performed to assess cell apoptosis. Our results showed pioglitazone pretreatment had a cytoprotective effect against H/R injury. The H/R + Pio group had an increased ratio of LC3-II to LC3-I and increased Beclin-1, decreased p62. Pioglitazone also reduced apoptosis and enhanced cell survival while inducing autophagy. Correspondingly, autophagy inhibition with 3-MA alleviated this protective effect. Furthermore, pioglitazone-induced enhancement of autophagy could be related to increased AMP-activated protein kinase (AMPK) phosphorylation and decreased Mammalian target of rapamycin (mTOR) phosphorylation. Thus, pioglitazone pretreatment protects against H/R injury by enhancting autophagy through the AMPK-mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Cong Zou
- Department of Endocrinology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Zhenfeng Ye
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Yawei Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Tongchang Chen
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, PR China.
| |
Collapse
|
29
|
ATG5-mediated autophagy suppresses NF-κB signaling to limit epithelial inflammatory response to kidney injury. Cell Death Dis 2019; 10:253. [PMID: 30874544 PMCID: PMC6420665 DOI: 10.1038/s41419-019-1483-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/14/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022]
Abstract
G2/M-arrested proximal tubular epithelial cells (TECs) after renal injury are linked to increased cytokines production. ATG5-mediated autophagy in proximal TECs has recently been shown to protect against G2/M cell cycle arrest and renal fibrosis. However, the impacts of autophagy in regulating inflammatorily response mounted by injured TECs remains largely unknown. In the present study, we investigated whether ATG5 acts as an innate immune suppressor in proximal TECs during kidney injury. Using the unilateral ureteric obstruction model in proximal tubule-specific autophagy-deficient mice, we demonstrated that ablation of epithelial ATG5 genes markedly impaired autophagy, resulting in enhanced nuclear factor κB (NF-κB) activation, macrophage and lymphocyte infiltration, and proinflammatory cytokines production in obstructed kidneys, as compared with wild-type mice. Following stimulation with angiotensin II (Ang II), siRNA silencing of ATG5 in cultured HK-2 cells or ATG5-deficient primary proximal TECs produced more cytokines, including IL-1β, IL-6, and TNF-α than did their control cells. Overexpressed ATG5, but not the autophagy-incompetent ATG5 mutant K130R in HK-2 cells, rendered resistant to Ang II-induced inflammatory response. Immunofluorescence assay indicated that ATG5 and p65 colocalized in the nucleus and cytoplasm, and their interaction was verified in immunoprecipitation assay from HEK-293T cell extracts. Genetic downregulation of endogenous ATG5 increased Ang II-induced phosphorylation and nuclear translocation of p65 and transcriptional activity of NF-κB, whereas the overexpressed ATG5, rather than ATG5 mutant K130R, hampered activation of NF-κB signaling, suggest an autophagy-dependent anti-inflammatory effect of ATG5. Further, pharmacological manipulation of autophagy yielded similar results both in vivo and in vitro. Additionally, JSH-23, a specific inhibitor of NF-κB nuclear translocation, rescued Ang II-driven IL-1β production in ATG5 siRNA-treated cells and decreased the proportion of cells in G2/M phase. In conclusion, ATG5-mediated autophagy in tubules targets NF-κB signaling to protect against renal inflammation.
Collapse
|
30
|
Zhang Z, Zhao H, Ge D, Wang S, Qi B. β-Casomorphin-7 Ameliorates Sepsis-Induced Acute Kidney Injury by Targeting NF-κB Pathway. Med Sci Monit 2019; 25:121-127. [PMID: 30610183 PMCID: PMC6330023 DOI: 10.12659/msm.912730] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background The aim of this study was to investigate the protective effect of β-casomorphin-7 (β-CM-7) and its possible mechanisms on acute kidney injury (AKI). Material/Methods Rats were randomly divided into a sham group, a cecal ligation and puncture (CLP) group, and a CLP+β-CM-7 group. Kidney index, kidney function, and histopathology changes were assessed. The expression of neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (Kim-1), nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα), and p-IκBα in kidney tissues were detected by Western blotting. Inflammatory and oxidative stress factors were detected by ELISA kits. Results The results showed that treatment with β-CM-7 reduced the levels of creatinine (Cre), blood urea nitrogen (BUN), NGAL, and Kim-1 induced by CLP, weakening the pathological damage. In the CLP + β-CM-7 group, the tumor necrosis factor-α (TNF-α) level and the DNA-binding activity of NF-κB p65 were significantly reduced and the interleukin-10 (IL-10) level was significantly increased compared with the CLP group. b-CM-7 decreased the expression of p-IκBα/IκBα. In addition, β-CM-7 increased the activity of superoxide dismutase (SOD) and decreased the level of malondialdehyde (MDA) in kidney tissue. Conclusions β-CM-7 attenuated sepsis-induced AKI through reducing inflammation and oxidative stress and by inhibition of nuclear factor (NF)-κB activities. This study provides a new therapeutic agent for attenuating sepsis-induced kidney injury.
Collapse
Affiliation(s)
- ZhiJie Zhang
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Huatang Zhao
- Department of Anesthesiology, Taishan Medical University Affiliated Liaocheng Second People's Hospital, Linqing, Shandong, China (mainland)
| | - DongJian Ge
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Shanshan Wang
- Department of Anesthesiology, Huaiyin Hospital of Huaian City, Huai'an, Jiangsu, China (mainland)
| | - Bin Qi
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| |
Collapse
|
31
|
Abstract
Inflammation is an adaptive process to the noxious stimuli that the human body is constantly exposed to. From the local inflammatory response to a full-blown systemic inflammation, a wide complex sequence of events occurs. Persistent immunosuppression and catabolism may ensue, until multiple organ failure finally sets in. And since clinically useful and specific biomarkers are lacking, diagnosis may come late. A thorough understanding of these events (how they begin, how they evolve, and how to modulate them) is imperative, but as yet poorly studied. This review aims to consolidate current knowledge of these events so that the management of these patients is not only evidence-based, but also built on an understanding of the inner workings of the human body in health and in disease.
Collapse
Affiliation(s)
- Miguel Lourenço Varela
- Internal Medicine 1, Hospital de Faro, Centro Hospitalar Universitário do Algarve, Rua Leão Penedo, Faro, 8000-386, Portugal.
- Intensive Care Medicine 1, Hospital de Faro, Centro Hospitalar Universitário do Algarve, Rua Leão Penedo, Faro, 8000-386, Portugal.
| | - Mihail Mogildea
- Internal Medicine 1, Hospital de Faro, Centro Hospitalar Universitário do Algarve, Rua Leão Penedo, Faro, 8000-386, Portugal
| | - Ignacio Moreno
- Internal Medicine 1, Hospital de Faro, Centro Hospitalar Universitário do Algarve, Rua Leão Penedo, Faro, 8000-386, Portugal
| | - Ana Lopes
- Internal Medicine 1, Hospital de Faro, Centro Hospitalar Universitário do Algarve, Rua Leão Penedo, Faro, 8000-386, Portugal
| |
Collapse
|
32
|
Aboutaleb N, Jamali H, Abolhasani M, Pazoki Toroudi H. Lavender oil (Lavandula angustifolia) attenuates renal ischemia/reperfusion injury in rats through suppression of inflammation, oxidative stress and apoptosis. Biomed Pharmacother 2018; 110:9-19. [PMID: 30453254 DOI: 10.1016/j.biopha.2018.11.045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/04/2018] [Accepted: 11/10/2018] [Indexed: 12/21/2022] Open
Abstract
Renal ischemia/reperfusion (I/R) injury following kidney transplantation has been found to be a great clinical problem owing to initiation of acute inflammatory responses and subsequently rapid loss of kidney function. It is well known that lavender oil exhibits an extensive spectrum of pharmacological and biochemical activities. The purpose of this study was to clarify molecular targets of lavender in treatment of this disease. Male Wistar rats weighing 200-250 g were divided into three major groups: sham, I/R, and I/R + different doses of lavender oil (L1:50 mg/kg, L2: 100 mg/kg, and L3: 200 mg/kg). A rat model of renal I/R (45 min ischemia and 24 h reperfusion) was created and lavender was administrated at 1 h after the beginning of reperfusion (i.p). Activities of antioxidant enzymes such as SOD, GPX, and CAT, and lipid peroxidation were evaluated. The expression of inflammatory cytokines such as TNFα, IL1β, and IL10 was determined by IHC and ELISA assay. Apoptosis activity and tissue damage were evaluated by TUNEL and H & E staining, respectively. Our results showed that lavender oil markedly restored activities of antioxidant enzymes and reduced lipid peroxidation (P < 0.05). Lavender significantly decreased levels of TNFα and IL1β and increased level of IL10 in a dose-dependent manner (P < 0.05). Lavender reduced TUNEL positive cells in a dose-dependent manner. However, lavender reduced damage to peritubular capillaries and contributed to preservation of normal morphology of renal cells. In sum, our findings establish a fundamental foundation for future drug industry to decrease the rates of rejection in kidney transplant patients.
Collapse
Affiliation(s)
- Nahid Aboutaleb
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hosein Jamali
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran; Pathology Department, Hasheminejad Kidney Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamidreza Pazoki Toroudi
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Sarhan M, Land WG, Tonnus W, Hugo CP, Linkermann A. Origin and Consequences of Necroinflammation. Physiol Rev 2018; 98:727-780. [PMID: 29465288 DOI: 10.1152/physrev.00041.2016] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When cells undergo necrotic cell death in either physiological or pathophysiological settings in vivo, they release highly immunogenic intracellular molecules and organelles into the interstitium and thereby represent the strongest known trigger of the immune system. With our increasing understanding of necrosis as a regulated and genetically determined process (RN, regulated necrosis), necrosis and necroinflammation can be pharmacologically prevented. This review discusses our current knowledge about signaling pathways of necrotic cell death as the origin of necroinflammation. Multiple pathways of RN such as necroptosis, ferroptosis, and pyroptosis have been evolutionary conserved most likely because of their differences in immunogenicity. As the consequence of necrosis, however, all necrotic cells release damage associated molecular patterns (DAMPs) that have been extensively investigated over the last two decades. Analysis of necroinflammation allows characterizing specific signatures for each particular pathway of cell death. While all RN-pathways share the release of DAMPs in general, most of them actively regulate the immune system by the additional expression and/or maturation of either pro- or anti-inflammatory cytokines/chemokines. In addition, DAMPs have been demonstrated to modulate the process of regeneration. For the purpose of better understanding of necroinflammation, we introduce a novel classification of DAMPs in this review to help detect the relative contribution of each RN-pathway to certain physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Walter G Land
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Wulf Tonnus
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Christian P Hugo
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Andreas Linkermann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
34
|
Cardiotrophin-1 Improves Kidney Preservation, Graft Function, and Survival in Transplanted Rats. Transplantation 2018; 102:e404-e412. [DOI: 10.1097/tp.0000000000002313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Chen W, Xi X, Zhang S, Zou C, Kuang R, Ye Z, Huang Y, Hu H. Pioglitazone Protects Against Renal Ischemia-Reperfusion Injury via the AMP-Activated Protein Kinase-Regulated Autophagy Pathway. Front Pharmacol 2018; 9:851. [PMID: 30127742 PMCID: PMC6088275 DOI: 10.3389/fphar.2018.00851] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a major cause of acute renal failure. Our previous studies have shown that pioglitazone, a peroxisome proliferators-activated receptor (PPAR)-γ agonist used in type 2 diabetes, protects against renal IRI; however, the molecular mechanism underlying the renoprotective effects of pioglitazone is still unclear. In this study, we investigated the role of AMP-activated protein kinase (AMPK)-regulated autophagy in renoprotection by pioglitazone in IRI. To investigate whether pioglitazone protects renal cells from IRI, an in vivo renal IRI model was used. Cell apoptosis in the kidneys was determined by TUNEL staining. Western blotting was used to determine the expression of AMPK, autophagy-related proteins, and caspase-3/8 proteins in the kidneys. In a rat model of IRI, pioglitazone decreased the increased serum creatinine and urea nitrogen, improved renal histological score, and decreased the cell injury. Pioglitazone also increased AMPK phosphorylation, inhibited p62 and cleaved caspase-3/8 proteins, and activated autophagy-related proteins LC3 II and Beclin-1 in the kidneys of IRI rats. Moreover, GW9662, as a selective inhibitor of PPAR-γ, inhibited the protective effects of pioglitazone. These results suggest that pioglitazone exerts its protective effects in renal IRI via activation of an AMPK-regulated autophagy signaling pathway.
Collapse
Affiliation(s)
- Wenlin Chen
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | | | - Cong Zou
- Department of Endocrinology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Renrui Kuang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenfeng Ye
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yawei Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
36
|
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: 20] [Impact Index Per Article: 2.9] [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
|
37
|
Chen H, Hu Y, Xie K, Chen Y, Wang H, Bian Y, Wang Y, Dong A, Yu Y. Effect of autophagy on allodynia, hyperalgesia and astrocyte activation in a rat model of neuropathic pain. Int J Mol Med 2018; 42:2009-2019. [PMID: 30015858 PMCID: PMC6108883 DOI: 10.3892/ijmm.2018.3763] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/04/2018] [Indexed: 02/05/2023] Open
Abstract
Primary damage or dysfunction of the nervous system may cause or initiate neuropathic pain. However, it has been difficult to establish an effective treatment for neuropathic pain, as the mechanisms responsible for its pathology remain largely unknown. Autophagy is closely associated with the pathological process of neurodegenerative diseases, neuropathic injury and cancer, among others. The aim of the present study was to examine the changes in the autophagy-lysosomal pathway and discuss the effects of autophagy on allodynia, hyperalgesia and astrocyte activation in neuropathic pain. A neuropathic pain model was induced by chronic constriction injury (CCI) in rats. Inducers and inhibitors of autophagy and lysosomes were used to assess autophagy, allodynia, hyperalgesia and astrocyte activity. Neuropathic pain was found to induce an increase in the levels of the autophagy-related proteins, LC3II and Beclin 1 and, and in those of the lysosomal proteins, lysosomal-associated membrane protein type 2 (LAMP2) and Ras-related protein Rab-7a (RAB7), whereas p62 levels were found to decrease from day 1 to 14 following CCI. The autophagy inducer, rapamycin, further increased the LC3II, Beclin 1, lysosomal-associated membrane protein 2 (LAMP2) and Ras-related protein Rab-7a (RAB7) expression levels, and decreased the p62 expression levels, which were accompanied by alleviation of allodynia, hyperalgesia and astrocyte activation in the rats subjected to CCI; the autophagy inhibitor, 3-methyladenine, reversed these effects. The use of the lysosomal inhibitors, bafilomycin and chloroquine, resulted in the accumulation of LC3II and Beclin 1, a decrease in the levels of LAMP2 and RAB7, and the exacerbation of allodynia, hyperalgesia and astrocyte activation in rats with neuropathic pain. On the whole, the findings of this study indicate that neuropathic pain activates autophagy, which alleviates mechanical and thermal hyperalgesia and suppresses astrocyte activity. Therefore, neuropathic pain induced by CCI in rats appears to be mediated via the autophagy-lysosomal pathway.
Collapse
Affiliation(s)
- Hongguang Chen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yajiao Hu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yajun Chen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Huixing Wang
- Pain Management Center, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yingxue Bian
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yanyan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Aili Dong
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
38
|
Gu L, Tao Y, Chen C, Ye Y, Xiong X, Sun Y. Initiation of the inflammatory response after renal ischemia/reperfusion injury during renal transplantation. Int Urol Nephrol 2018; 50:2027-2035. [PMID: 29974405 DOI: 10.1007/s11255-018-1918-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/20/2018] [Indexed: 01/22/2023]
Abstract
Ischemia/reperfusion injury (IRI) occurs commonly during renal transplantation. It has been well demonstrated that the inflammatory response has an important role in the pathogenesis and pathological processes of IRI. However, the signaling events that trigger the activation of the inflammatory response are less clear. Accumulated evidence has identified the role of various injury factors released from or exposed in ischemic, damaged, or dying cells, which serve as initiators of the inflammatory response and exacerbate kidney injury after renal IRI. Signaling pathways triggered by these endogenous molecules that activate different pathogen recognition receptors have also been widely investigated. Here, we review the molecular signaling molecules that initiate the inflammatory response during renal IRI and that provide potential therapeutic options for the disease.
Collapse
Affiliation(s)
- Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Tao
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Cheng Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yao Sun
- Key Laboratory of Pesticides and Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, Hubei International Scientific and Technology Cooperation Base of Pesticide and Green Synthesis, Chemical Biology Center, College of Chemistry, Central China Normal Universtiy, Wuhan, 430079, China.
| |
Collapse
|
39
|
Hadj Abdallah N, Baulies A, Bouhlel A, Bejaoui M, Zaouali MA, Ben Mimouna S, Messaoudi I, Fernandez-Checa JC, García Ruiz C, Ben Abdennebi H. The effect of zinc acexamate on oxidative stress, inflammation and mitochondria induced apoptosis in rat model of renal warm ischemia. Biomed Pharmacother 2018; 105:573-581. [PMID: 29890465 DOI: 10.1016/j.biopha.2018.06.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
AIM Zinc has proved its efficacy in many models of ischemia reperfusion (I/R) injury. In this study, we used zinc acexamate (ZAC) as an exogenous source of zinc against renal I/R injury and we investigated whether its protective effects are mediated by the decrease of oxidative stress, inflammation, and mitochondria induced-apoptosis. METHODS Rats were orally pretreated with vehicle or ZAC (10 or 100 mg/kg) 24 h and 30 min prior to 1 h of bilateral renal warm ischemia and 2 h of reperfusion. RESULTS Our data showed that 10 mg/kg of ZAC, but not 100 mg/kg, improved renal architecture and function. Also, the low dose of ZAC up-regulated antioxidant enzymes activities and glutathione level and decreased lipids and proteins oxidation. Interestingly, the use of ZAC resulted in a significant reduce of pro-inflammatory cytokines (IL-1ß, IL-6 and MCP-1), enhanced mitochondria integrity and decreased expression of the pro-apoptotic protein caspase-9. CONCLUSION We conclude that renal I/R induced oxidative stress, inflammation and apoptosis and that the use of ZAC at 10 mg/kg, but not 100 mg/kg, protects rat kidneys from I/R injury by down-regulating these processes.
Collapse
Affiliation(s)
- Najet Hadj Abdallah
- Department of physiology, Unité de Biologie et anthropologie moléculaires appliquées au développement et à la santé, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| | - Anna Baulies
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas. Liver Unit Hospital Clínici Provincial, IDIBAPS and CIBERehd, 08036, Barcelona, Spain.
| | - Ahlem Bouhlel
- Department of physiology, Unité de Biologie et anthropologie moléculaires appliquées au développement et à la santé, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| | - Mohamed Bejaoui
- Department of physiology, Unité de Biologie et anthropologie moléculaires appliquées au développement et à la santé, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| | - Mohamed Amine Zaouali
- Department of physiology, Unité de Biologie et anthropologie moléculaires appliquées au développement et à la santé, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| | - Safa Ben Mimouna
- Laboratoire de Génétique, Biodiversité et Valorisation des Bioressources (LR11ES41). Institute of Biotechnology, University of Monastir, Monastir, Tunisia.
| | - Imed Messaoudi
- Laboratoire de Génétique, Biodiversité et Valorisation des Bioressources (LR11ES41). Institute of Biotechnology, University of Monastir, Monastir, Tunisia.
| | - José Carlos Fernandez-Checa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas. Liver Unit Hospital Clínici Provincial, IDIBAPS and CIBERehd, 08036, Barcelona, Spain.
| | - Carmen García Ruiz
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas. Liver Unit Hospital Clínici Provincial, IDIBAPS and CIBERehd, 08036, Barcelona, Spain.
| | - Hassen Ben Abdennebi
- Department of physiology, Unité de Biologie et anthropologie moléculaires appliquées au développement et à la santé, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| |
Collapse
|
40
|
Gong J, Wang X. Schisantherin A protects renal tubular epithelial cells from hypoxia/reoxygenation injury through the activation of PI3K/Akt signaling pathway. J Biochem Mol Toxicol 2018; 32:e22160. [PMID: 29785781 DOI: 10.1002/jbt.22160] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 04/24/2018] [Accepted: 05/03/2018] [Indexed: 01/10/2023]
Abstract
Schisantherin A (SchA), a dibenzocyclooctadiene lignan isolated from the fruit of Schisandra sphenanthera, was reported to possess anti-inflammatory and antioxidant activities. However, its protective effect against renal ischemia-reperfusion (I/R) injury in human renal tubular epithelial cells subjected to hypoxia/reoxygenation (H/R) has never been studied. Thus, herein, we investigated the effect of SchA on renal I/R injury in vitro. Our results demonstrated that SchA pretreatment significantly improved HK-2 cell viability exposed to H/R. Pretreatment with SchA markedly inhibited the levels of reactive oxygen species and malondialdehyde, as well as suppressed the production of tumor necrosis factor-α (TNF-α), interleukin-1β, and interleukin-6 in H/R-stimulated HK-2 cells. In addition, SchA also suppressed H/R-induced HK-2 cell apoptosis. Furthermore, this protective effect of SchA was mediated through the PI3K/Akt signaling pathway in HK-2 cells. These findings showed that SchA may exert a protective effect on renal tubular epithelial cells against H/R injury through the activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Jiachuan Gong
- Department of Nephrology and Rheumatology, The First People's Hospital of Shangqiu, Shangqiu, 476100, Henan, People's Republic of China
| | - Xuezhen Wang
- Department of Nephrology and Rheumatology, The First People's Hospital of Shangqiu, Shangqiu, 476100, Henan, People's Republic of China
| |
Collapse
|
41
|
Wu M, Hu M, Tong H, Liu J, Jiang H, Zhang M, Su L, Li M, Feng Y, Cheng B. Regulatory mechanism of ulinastatin on autophagy of macrophages and renal tubular epithelial cells. OPEN CHEM 2018. [DOI: 10.1515/chem-2018-0025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Kidney ischemia and hypoxia can cause renal cell apoptosis and activation of inflammatory cells, which lead to the release of inflammatory factors and ultimately result in the damage of kidney tissue and the whole body. Renal tubular cell and macrophage autophagy can reduce the production of reactive oxygen species (ROS), thereby reducing the activation of inflammatory cytoplasm and its key effector protein, caspase-1, which reduces the expression of IL-1β and IL-18 and other inflammatory factors. Ulinastatin (UTI), as a glycoprotein drug, inhibits the activity of multiple proteases and reduces myocardial damage caused by ischemia-reperfusion by upregulating autophagy. However, it can be raised by macrophage autophagy, reduce the production of ROS, and ultimately reduce the expression of inflammatory mediators, thereby reducing renal cell injury, promote renal function recovery is not clear. In this study, a series of cell experiments have shown that ulinastatin is reduced by regulating the autophagy of renal tubular epithelial cells and macrophages to reduce the production of reactive oxygen species and inflammatory factors (TNF-α, IL-1β and IL-1), and then, increase the activity of the cells under the sugar oxygen deprivation model. The simultaneous use of cellular autophagy agonists Rapamycin (RAPA) and ulinastatin has a synergistic effect on the production of reactive oxygen species and the expression of inflammatory factors.
Collapse
Affiliation(s)
- Ming Wu
- Department of Critical Care Medicine , ShenZhen No.2 People’s Hospital , Shenzhen 518035 , China
- Southern Medical University , Guangzhou 510515 , China
| | - Min Hu
- Emergency Department, Changhai Hospital , Shanghai 200433 , China
| | - Huansheng Tong
- Department of Intensive Care Unit, Guangzhou School of Clinical Medicine , Southern Medical University (Guangzhou General Hospital of Guangzhou Military Region) , Guangzhou 510010 , China
| | - Junying Liu
- Department of Endocrinology , Shen Zhen No.2 People’s Hospital , Shenzhen 518035 , China
| | - Hui Jiang
- Department of Critical Care Medicine , ShenZhen No.2 People’s Hospital , Shenzhen 518035 , China
| | - Ming Zhang
- Southern Medical University , Guangzhou 510515 , China
- Department of Critical Care Medicine , The People’ Hospital of Qin Yuan , Qin Yuan 511500 , China
| | - Lei Su
- Department of Intensive Care Unit, Guangzhou School of Clinical Medicine , Southern Medical University (Guangzhou General Hospital of Guangzhou Military Region) , Guangzhou 510010 , China
| | - Mingli Li
- Department of interventional therapy , Shen Zhen No.2 People’s Hospital , Shenzhen 518035 , P.R. China
| | - Yongwen Feng
- Department of Critical Care Medicine , ShenZhen No.2 People’s Hospital , Shenzhen 518035 , China
| | - Biao Cheng
- Department of Plastic Surgery, Guangzhou School of Clinical Medicine , Southern Medical University (Guangzhou General Hospital of Guangzhou Military Region) , Guangzhou 510010 , China
| |
Collapse
|
42
|
Amirzargar MA, Yaghubi F, Hosseinipanah M, Jafari M, Pourjafar M, Rezaeepoor M, Rezaei H, Roshanaei G, Hajilooi M, Solgi G. Anti-inflammatory Effects of Valproic Acid in a Rat Model of Renal Ischemia/Reperfusion Injury: Alteration in Cytokine Profile. Inflammation 2018; 40:1310-1318. [PMID: 28478517 DOI: 10.1007/s10753-017-0574-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Valporic acid (VPA) has been implicated to have anti-inflammatory and anti-oxidant activities in several ischemic/reperfusion (I/R) injury models. This study intended to evaluate whether VPA could affect the inflammatory/anti-inflammatory cytokines balance and severity of renal I/R injury in rat. I/R injury was induced in two groups of animals, vehicle normal saline and VPA-treated (IP injection, 150 mg/kg) rats, by 45 min occlusion of both left and right renal arteries followed by 3, 24 and 120 h reperfusion in separate groups. After each time point, kidneys and blood samples were collected for cytokine genes (TNF-α, IL-1β, IL-10 and TGF-β) expression analysis and histological examinations in the kidney tissues. Serum creatinine levels were measured for evaluation of renal function. We observed significantly downregulated mRNA expressions for IL-1β and TNF-α in blood and tissue samples 24 and 120 h post I/R injury in VPA-treated animals compared to control groups (P < 0.0001). On the other hand, mRNA expression levels for IL-10 and TGF-β were significantly increased in the blood samples from VPA-treated animals at two time points after I/R injury (P < 0.0001) and at 120 h in tissue samples (P < 0.001). Histopathology analysis showed downgraded ischemic changes in VPA group compared to sham control. Also, decreased serum creatinine levels were observed in VPA-treated animals particularly 120 h post I/R injury (P < 0.0001) that was correlated with less pathological changes in this group. Our results indicate that VPA can attenuate pro-inflammatory responses and augment the anti-inflammatory condition in favor of faster renal recovery from ischemic changes and improved renal function after renal I/R injury.
Collapse
Affiliation(s)
- Mohammad Ali Amirzargar
- Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Faramarz Yaghubi
- Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Hosseinipanah
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Jafari
- Department of Pathology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Pourjafar
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Rezaeepoor
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamzeh Rezaei
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Godratollah Roshanaei
- Research Center for Health Science, Department of Biostatistics and Epidemiology, School of Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrdad Hajilooi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
43
|
Nanoparticle-mediated dual delivery of resveratrol and DAP5 ameliorates kidney ischemia/reperfusion injury by inhibiting cell apoptosis and inflammation. Oncotarget 2018; 8:39547-39558. [PMID: 28465474 PMCID: PMC5503631 DOI: 10.18632/oncotarget.17135] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/12/2017] [Indexed: 12/30/2022] Open
Abstract
Ischemia reperfusion (I/R) injury is a leading cause of acute kidney injury with high morbidity and mortality due to limited therapy. NMDA receptor inhibitor (DAP5) and resveratrol (Res) could ameliorate kidney I/R injury, but their use is limited by low aqueous solubility and poor stability. Here, we examined the potential protective effects of Res-DAP5 nanoparticles (NP) against renal I/R injury. Mice were subjected to renal ischemia for 30 min followed by reperfusion for 24 h. The results showed that Res-DAP5-NP could decreased serum creatinine (Cr) and urea nitrogen (BUN), alleviated tubular damage and oxidative stress. In addition, Res-DAP5-NP suppressed cell apoptosis, promoted the expression of p-DAPK, and inhibited the expression of p-CaMK and p-AKT. Furthermore, Res-DAP5-NP decreased the production of pro-inflammatory cytokines such as tumor necrosis factor-α, IL-1β, IL-6, and p-IκBα induced by renal I/R injury. In addition, Res-DAP5-NP also attenuated renal I/R injury in vivo, as manifested by increase in cell viability, SOD level, and the expression of p-DAPK, decreases in intracellular Ca2+ concentration and the expression of p-CaMK. Taken together, our findings indicates that Res-DAP5-NP could effectively protect renal I/R injury by inhibiting apoptosis and inflammation responses, possibly through AKT/NMDA/CaMK/DAPK and NF-κB pathways.
Collapse
|
44
|
Jia H, Yan Y, Liang Z, Tandra N, Zhang B, Wang J, Xu W, Qian H. Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review). Mol Med Rep 2018; 17:3439-3447. [PMID: 29257336 DOI: 10.3892/mmr.2017.8311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/09/2017] [Indexed: 11/09/2022] Open
Abstract
Acute kidney injury (AKI) is a common and serious medical condition associated with poor health outcomes. Autophagy is a conserved multistep pathway that serves a major role in many biological processes and diseases. Recent studies have demonstrated that autophagy is induced in proximal tubular cells during AKI. Autophagy serves a pro‑survival or pro‑death role under certain conditions. Furthermore, mesenchymal stem cells (MSCs) have therapeutic potential in the repair of renal injury. This review summarizes the recent progress on the role of autophagy in AKI and MSCs‑based therapy for AKI. Further research is expected to prevent and treat acute kidney injury.
Collapse
Affiliation(s)
- Haoyuan Jia
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yongmin Yan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaofeng Liang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Nitin Tandra
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Bin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Juanjuan Wang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
45
|
Zhang D, Zhou J, Ye LC, Li J, Wu Z, Li Y, Li C. Autophagy maintains the integrity of endothelial barrier in LPS-induced lung injury. J Cell Physiol 2018; 233:688-698. [PMID: 28328069 DOI: 10.1002/jcp.25928] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/21/2017] [Indexed: 12/26/2022]
Abstract
Understanding the role and underlying regulation mechanism of autophagy in lipopolysaccharide-induced lung injury (LPS-LI) may provide potentially new pharmacological targets for treatment of acute lung injury. The aim of this study was to investigate the functional significance of autophagy in LPS-LI. The autophagy of human pulmonary microvascular endothelial cells (HPMVECs) and mice was inhibited before they were challenged with LPS. In vitro, permeability, vitality, and the LDH release rate of the cells were detected, the zonula occluden-1 (ZO-1) expression and the stress fiber formation were determined. In vivo, the lung injury was assessed. We found LPS caused high permeability and increased lactate dehydrogenase (LDH) release rate, lowered viability of the cells, inhibited the ZO-1 expression and induced stress fiber formation, these effects were further aggravated by prohibiting the level of autophagy. Consistently, in in vivo experiments, LPS-induced serious lung injury, which was reflected as edema, leukocyte infiltration and hemorrhage in lung tissue, and the high concentration of pro-inflammation cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-1β in bronchoalveolar lavage fluid (BALF). Inhibiting autophagy further exacerbated LPS-LI. It appears that autophagy played a protective role in LPS-LI in part through restricting the injury of lung microvascular barrier.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, P.R. China
| | - Jian Zhou
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Le Chi Ye
- Department of Oncological Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, P.R. China
| | - Jing Li
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Zhenzhou Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, P.R. China
| | - Yuping Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, P.R. China
| | - Chichi Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, P.R. China
| |
Collapse
|
46
|
Wang C, Wang Q, Lou Y, Xu J, Feng Z, Chen Y, Tang Q, Zheng G, Zhang Z, Wu Y, Tian N, Zhou Y, Xu H, Zhang X. Salidroside attenuates neuroinflammation and improves functional recovery after spinal cord injury through microglia polarization regulation. J Cell Mol Med 2017; 22:1148-1166. [PMID: 29148269 PMCID: PMC5783886 DOI: 10.1111/jcmm.13368] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/03/2017] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disease; however, few drugs have been proved to treat SCI effectively. Neuroinflammation is the major pathogenesis of SCI secondary injury and considered to be the therapeutic target of SCI. Salidroside (Sal) has been reported to exert anti‐inflammatory effects in airway, adipose and myocardial tissue; however, the role of Sal in SCI therapeutics has not been clarified. In this study, we showed that Sal could improve the functional recovery of spinal cord in rats as revealed by increased BBB locomotor rating scale, angle of incline, and decreased cavity of spinal cord injury and apoptosis of neurons in vivo. Immunofluorescence double staining of microglia marker and M1/M2 marker demonstrated that Sal could suppress M1 microglia polarization and activate M2 microglia polarization in vivo. To verify how Sal exerts its effects on microglia polarization and neuron protection, we performed the mechanism study in vitro in microglia cell line BV‐2 and neuron cell line PC12. The results showed that Sal prevents apoptosis of PC12 cells in coculture with LPS‐induced M1 BV‐2 microglia, also the inflammatory secretion phenotype of M1 BV‐2 microglia was suppressed by Sal, and further studies demonstrated that autophagic flux regulation through AMPK/mTOR pathway was involved in Sal regulated microglia polarization after SCI. Overall, our study illustrated that Sal could promote spinal cord injury functional recovery in rats, and the mechanism may relate to its microglia polarization modulation through AMPK‐/mTOR‐mediated autophagic flux stimulation.
Collapse
Affiliation(s)
- Chenggui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Qingqing Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yiting Lou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Jianxiang Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Zhenhua Feng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Qian Tang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Gang Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Zengjie Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yaosen Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
| |
Collapse
|
47
|
Wu X, Yan T, Wang Z, Wu X, Cao G, Zhang C, Tian X, Wang J. Micro-vesicles derived from human Wharton's Jelly mesenchymal stromal cells mitigate renal ischemia-reperfusion injury in rats after cardiac death renal transplantation. J Cell Biochem 2017; 119:1879-1888. [PMID: 28815768 DOI: 10.1002/jcb.26348] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/11/2017] [Indexed: 01/12/2023]
Abstract
The purpose of the present study was to investigate the possible therapeutic effects of the human Wharton-Jelly mesenchymal stromal cells derived micro-vesicles (hWJMSCs-MVs) on renal ischemia-reperfusion injury (IRI) after cardiac death (CD) renal transplantation in rats. MVs were injected intravenously in rats immediately after renal transplantation. The animals were sacrificed at 24 h, 48 h, 1 and 2 weeks post-transplantation. ELISA was used to determine the von Willebrand Factor (vWF), tumor necrosis factor (TNF)-α, and interleukin (IL)-10 levels in the serum. Tubular cell proliferation and apoptosis were identified by Ki67 immunostaining and TUNEL assay. Renal fibrosis was assessed by Masson's tri-chrome straining and alpha-smooth muscle actin (α-SMA) staining. The infiltration of inflammatory cells was detected by CD68+ staining. The transforming growth factor (TGF)-β, hepatocyte growth factor (HGF), and α-SMA expression in the kidney was measured by Western blot. After renal transplantation, the rats treated with hWJMSCs-MVs improved survival rate and renal function. Moreover, MVs mitigated renal cell apoptosis, enhanced proliferation, and alleviated inflammation at the first 48 h. In the late period, abrogation of renal fibrosis was observed in the MVs group. MVs also could decrease the number of CD68+ macrophages in the kidney. Furthermore, MVs decreased the protein expression levels of α-SMA and TGF-β1 and increased the protein expression level of HGF at any point (24 h, 48 h, 1 or 2 weeks). The administration of MVs immediately after renal transplantation could ameliorate IRI in both the acute and chronic stage.
Collapse
Affiliation(s)
- Xiaoqiang Wu
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Tianzhong Yan
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhiwei Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xuan Wu
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Guanghui Cao
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Chan Zhang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiangyong Tian
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Junpeng Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
48
|
The mTOR signal regulates myeloid-derived suppressor cells differentiation and immunosuppressive function in acute kidney injury. Cell Death Dis 2017; 8:e2695. [PMID: 28333137 PMCID: PMC5386577 DOI: 10.1038/cddis.2017.86] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
The mammalian target of rapamycin (mTOR) signal controls innate and adaptive immune response in multiple immunoregulatory contexts. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid cells of potent immunosuppressive capacity. In this study, we aimed to investigate the role of MDSCs in the protection of acute kidney injury (AKI) and the regulation of mTOR signal on MDSC's protective role in this context. In mice AKI model, rapamycin administration was associated with improved renal function, restored histological damage and decreased CD4+ and CD8+ T-cell infiltration in kidney tissue. MDSCs, especially CD11b+Ly6G+Ly6Clow G-MDSCs were recruited to the injured kidney following the interaction of CXCL1, CXCL2 and their receptor CXCR2 after inhibiting mTOR signal with rapamycin treatment. The adoptive transfer of rapamycin-treated MDSCs into the mice with AKI significantly improved the renal function, ameliorated histologic damages and limited the infiltration of T cells in kidney tissue. In addition, the expression of pro-inflammatory cytokines IL-1β and IFN-γ mRNA was downregulated while the expression of TGF-β1 and Foxp3 mRNA was upregulated in kidney tissue after transferring rapamycin-treated MDSCs. Adoptive transfer of rapamycin-treated MDSCs also downregulated the serum levels of IL-1β, IL-6 and IFN-γ and upregulated the serum levels of TGF-β1 compared with the IR group and PBS-treated MDSC group. In in vitro study, inhibiting mTOR signal regulated the induction of MDSC towards the CD11b+Ly6G+Ly6Clow G-MDSC subset. The ability to suppress T-cell proliferation of both bone marrow–derived CD11b+Ly6G+Ly6Clow G-MDSCs and CD11b+Ly6G-Ly6Chigh M-MDSCs was enhanced by mTOR signal inhibition via upregulating the expression of Arginase-1 and iNOS. Accordingly, both G-MDSCs and M-MDSCs presented downregulated runx1 gene expression after rapamycin treatment. Taken together, our results demonstrated that MDSCs ameliorated AKI and the protective effect was enhanced by mTOR signal inhibition via promoting MDSCs recruitment, regulating the induction of MDSCs and strengthening their immunosuppressive activity.
Collapse
|
49
|
Yu A, Zhang T, Zhong W, Duan H, Wang S, Ye P, Wang J, Zhong S, Yang Z. miRNA-144 induces microglial autophagy and inflammation following intracerebral hemorrhage. Immunol Lett 2017; 182:18-23. [PMID: 28062218 DOI: 10.1016/j.imlet.2017.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/31/2016] [Accepted: 01/02/2017] [Indexed: 12/21/2022]
Abstract
Autophagic activation mediated inflammation contributes to brain injury of intracerebral hemorrhage (ICH). MiRNAs play a key role in inflammation, which negatively and posttranscriptionally regulate gene expression and function. Modulating the mTOR signal, a central regulator of autophagy, could be of great significance for ICH. However, the specific of miRNA is unknown. In the current study, we detected the miRNA-144 expression, autophagic activity and inflammation of microglia in ICH. We also knocked down endogenous miRNA-144 to regulate autophagy and inflammation in ICH. In addition, we assessed the neurological damge in ICH mice. We found that ICH promoted miRNA-144 expression but downregulated mTOR expression. In addition, upregulation of mTOR attenuated microglial autophagy and inflammation in ICH. Furthermore, downregulation of miRNA-144 also inhibited inflammation, brain edema and improved neurological functions in ICH mice. Taken together, our findings suggested that miRNA-144 was a crucial regulator of autophagy via regulation of mTOR, and represented a promising therapeutical strategy for ICH.
Collapse
Affiliation(s)
- Anyong Yu
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Tianxi Zhang
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Wenyi Zhong
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Haizhen Duan
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Song Wang
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Peng Ye
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Juan Wang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Shanchuan Zhong
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zhao Yang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
50
|
Lekli I, Haines DD, Balla G, Tosaki A. Autophagy: an adaptive physiological countermeasure to cellular senescence and ischaemia/reperfusion-associated cardiac arrhythmias. J Cell Mol Med 2016; 21:1058-1072. [PMID: 27997746 PMCID: PMC5431132 DOI: 10.1111/jcmm.13053] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/04/2016] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress placed on tissues that involved in pathogenesis of a disease activates compensatory metabolic changes, such as DNA damage repair that in turn causes intracellular accumulation of detritus and ‘proteotoxic stress’, leading to emergence of ‘senescent’ cellular phenotypes, which express high levels of inflammatory mediators, resulting in degradation of tissue function. Proteotoxic stress resulting from hyperactive inflammation following reperfusion of ischaemic tissue causes accumulation of proteinaceous debris in cells of the heart in ways that cause potentially fatal arrhythmias, in particular ventricular fibrillation (VF). An adaptive response to VF is occurrence of autophagy, an intracellular bulk degradation of damaged macromolecules and organelles that may restore cellular and tissue homoeostasis, improving chances for recovery. Nevertheless, depending on the type and intensity of stressors and inflammatory responses, autophagy may become pathological, resulting in excessive cell death. The present review examines the multilayered defences that cells have evolved to reduce proteotoxic stress by degradation of potentially toxic material beginning with endoplasmic reticulum‐associated degradation, and the unfolded protein response, which are mechanisms for removal from the endoplasmic reticulum of misfolded proteins, and then progressing through the stages of autophagy, including descriptions of autophagosomes and related vesicular structures which process material for degradation and autophagy‐associated proteins including Beclin‐1 and regulatory complexes. The physiological roles of each mode of proteotoxic defence will be examined along with consideration of how emerging understanding of autophagy, along with a newly discovered regulatory cell type called telocytes, may be used to augment existing strategies for the prevention and management of cardiovascular disease.
Collapse
Affiliation(s)
- Istvan Lekli
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - David Donald Haines
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Balla
- Department of Pediatrics, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary.,Hemostasis, Thrombosis and Vascular Biology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary
| | - Arpad Tosaki
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|