1
|
Nam D, Park J, Lee J, Son J, Kim JE. mTOR potentiates senescent phenotypes and primary cilia formation after cisplatin-induced G2 arrest in retinal pigment epithelial cells. Cell Signal 2024; 124:111402. [PMID: 39251051 DOI: 10.1016/j.cellsig.2024.111402] [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: 02/27/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cisplatin, a platinum-based anticancer drug, is used to treat several types of cancer. Despite its effectiveness, cisplatin-induced side effects have often been reported. Although cisplatin-induced toxicities, such as apoptosis and/or necrosis, have been well studied, the fate of cells after exposure to sublethal doses of cisplatin needs further elucidation. Treatment with a sublethal dose of cisplatin induced cell cycle arrest at the G2 phase in retinal pigment epithelial cells. Following cisplatin withdrawal, the cells irreversibly exited the cell cycle and became senescent. Notably, the progression from the G2 to the G1 phase occurred without mitotic entry, a phenomenon referred to as mitotic bypass, resulting in the accumulation of cells containing 4N DNA content. Cisplatin-exposed cells exhibited morphological changes associated with senescence, including an enlarged size of cell and nucleus and increased granularity. In addition, the senescent cells possessed primary cilia and persistent DNA lesions. Senescence induced by transient exposure to cisplatin involves mTOR activation. Although transient co-exposure with an mTORC1 inhibitor rapamycin did not prevent mitotic bypass and entry into senescence, it delayed the progression of senescence and attenuated senescent phenotypes, resulting in shorter primary cilia formation. Conclusively, cisplatin induces senescence in retinal pigment epithelial cells by promoting mTOR activation.
Collapse
Affiliation(s)
- Dajeong Nam
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaejung Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaehong Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juyoung Son
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
2
|
Li Z, Liu H, Xie Q, Yin G. Macrophage involvement in idiopathic inflammatory myopathy: pathogenic mechanisms and therapeutic prospects. J Inflamm (Lond) 2024; 21:48. [PMID: 39593038 PMCID: PMC11590228 DOI: 10.1186/s12950-024-00422-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Idiopathic inflammatory myopathies are a group of systemic autoimmune diseases characterized by chronic muscle inflammation and diverse clinical manifestations. Macrophages, pivotal components of innate immunity, are implicated in immune responses, inflammation resolution, and tissue repair. Distinct macrophage polarization states play vital roles in disease progression and resolution. Mechanistically, activated macrophages release proinflammatory cytokines, chemokines, and reactive oxygen species, perpetuating immune responses and tissue damage. Dysregulated macrophage polarization contributes to sustained inflammation. Here, we reviewed the intricate contributions of macrophages to IIM pathogenesis and explored novel therapeutic avenues. We discussed emerging strategies targeting macrophages, including receptor-based interventions and macrophage polarization modulation, for IIM treatment. This review underscores the multifaceted involvement of macrophages in IIM pathogenesis and offers insights into potential therapeutic approaches targeting these immune cells for disease management.
Collapse
Affiliation(s)
- Ziqi Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China
| | - Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China
| | - Geng Yin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China.
- Department of General Practice, West China Hospital, General Practice Medical Center, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Wang J, Du J, Gou X, Huang Y, He J, Lu X, Xie M. Propyl acetate protects intestinal barrier during parenteral nutrition in mice and Caco-2 cells. JPEN J Parenter Enteral Nutr 2024; 48:917-926. [PMID: 39187914 DOI: 10.1002/jpen.2681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Gut microbiota dysbiosis induces intestinal barrier damage during parenteral nutrition (PN). However, the underlying mechanisms remain unclear. This study aimed to investigate gut microbiota dysbiosis, luminal short-chain fatty acids, and autophagy in a mouse model and how these short-chain fatty acids regulate autophagy. METHODS Eight-week-old male specific-pathogen-free mice were randomly divided into a Chow group (standard diet and intravenous normal saline infusion) and a PN group (continuous infusion of PN nutrient solution) for 7 days. Caco-2 cells were also treated with intestinal rinse solutions from Chow and PN mouse models. RESULTS Compared with the Chow group, the PN group exhibited increased Proteobacteria and decreased Firmicutes, correlating with decreased propyl acetate. In the PN group, intestinal tissue exhibited elevated adenosine monophosphate-activated protein kinase (AMPK) phosphorylation, LC3II protein levels, and Atg3 and Atg7 messenger RNA levels. P62 protein levels were decreased, indicating an increase of autophagy flux in the PN group. In the Caco-2 cell model, cells treated with PN solution plus propyl acetate exhibited increased Claudin-1 and occluding along with decreased interleukin-6 and tumor necrosis factor α compared with those treated with PN solution alone. Propyl acetate addition inhibited the AMPK-mammalian target of rapamycin (mTOR) pathway, mitigating the excessive autophagy induced by the PN intestinal rinse solution in Caco-2 cells. CONCLUSION PN led to a significant reduction in propyl acetate levels in the intestine, excessive activation of autophagy, and barrier dysfunction. Propyl acetate inhibited excessive autophagy via the AMPK/mTOR signaling pathway and protected the intestinal barrier during PN.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jing Du
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaomei Gou
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yong Huang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jixin He
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoyun Lu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ming Xie
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
4
|
Zhang H, Deng Z, Wang Y, Zheng X, Zhou L, Yan S, Wang Y, Dai Y, Kanwar YS, Chen F, Deng F. CHIP drives proteasomal degradation of NUR77 to alleviate oxidative stress and intrinsic apoptosis in cisplatin-induced nephropathy. Commun Biol 2024; 7:1403. [PMID: 39462094 PMCID: PMC11513124 DOI: 10.1038/s42003-024-07118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Carboxy-terminus of Hsc70-interacting protein (CHIP), an E3 ligase, modulates the stability of its targeted proteins to alleviate various pathological perturbations in various organ systems. Cisplatin is a widely used chemotherapeutic agent, but it is also known for its alarming renal toxicity. The role of CHIP in the pathogenesis of cisplatin-induced acute kidney injury (AKI) has not been adequately investigated. Herein, we demonstrated that CHIP was abundantly expressed in the renal proximal tubular epithelia, and its expression was downregulated in cisplatin-induced AKI. Further investigation revealed that CHIP overexpression or activation alleviated, while its gene disruption promoted, oxidative stress and apoptosis in renal proximal tubular epithelia induced by cisplatin. In terms of mechanism, CHIP interacted with and ubiquitinated NUR77 to promote its degradation, which consequently shielded BCL2 to maintain mitochondrial permeability of renal proximal tubular cells in the presence of cisplatin. Also, we demonstrated that CHIP interacted with NUR77 via its central coiled-coil (CC) domain, a non-canonical interactive pattern. In conclusion, these findings indicated that CHIP ubiquitinated and degraded its substrate NUR77 to attenuate intrinsic apoptosis in cisplatin-treated renal proximal tubular epithelia, thus providing a novel insight for the pathogenesis of cisplatin-induced AKI.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zebin Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Yilong Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Zheng
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Lizhi Zhou
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Shu Yan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Yashpal S Kanwar
- Departments of Pathology & Medicine, Northwestern University, Chicago, IL, USA
| | - Fangzhi Chen
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
5
|
Barmaki H, Nourazarian A, Shademan B, Khaki-Khatibi F. The autophagy paradox: A new hypothesis in neurodegenerative disorders. Neurochem Int 2024; 179:105827. [PMID: 39111406 DOI: 10.1016/j.neuint.2024.105827] [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: 05/22/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
A recent study showed that while autophagy is usually tied to protein and organelle turnover, it can also play dual roles in neurodegenerative diseases. Traditionally, autophagy was seen as protective since it removes damaged proteins and organelles. but new data suggests autophagy can sometimes promote neuron death. and This review tackles autophagy's seemingly contradictory effects in neurodegeneration, or the "autophagy paradox. " It offers a framework for understanding autophagy in neurodegenerative research and the cellular processes involved. In short, our data uncovers a harmful autophagy role in certain situations, conflicting the view that it's always beneficial. We describe the distinct, disease-specific autophagy pathways functioning in various neurodegenerative diseases. Part two concerns potential therapeutic implications of manipulating autophagy and current strategies targeting the autophagic system, suggesting interesting areas for future research into tailored modulators. This could eventually enable activating or controlling specific autophagy pathways and aid in developing more effective treatments. Researchers believe more molecular-level research is needed so patient-tailored autophagy-modulating therapeutics can be developed given this dilemma. Moreover, research must translate faster into effective neurodegenerative disease treatment options. This article aims to provide a wholly new perspective on autophagy's classically described role in these severe diseases, challenging current dogma and opening new therapeutic avenue options.
Collapse
Affiliation(s)
- Haleh Barmaki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran; Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran.
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Dou L, Liu C, Su R, Corazzin M, Jin Z, Yang Z, Hu G, Zhang M, Sun L, Zhao L, Jin Y, Su L. Effects of dietary arginine supplementation on muscle structure, meat characteristics and lipid oxidation products in lambs and its potential mechanisms of action. Meat Sci 2024; 216:109581. [PMID: 38970933 DOI: 10.1016/j.meatsci.2024.109581] [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: 01/22/2024] [Revised: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
This study aimed to assess the effect of dietary arginine supplementation on muscle structure and meat characteristics of lambs also considering lipid oxidation products and to contribute to reveal its mechanisms of action using tandem mass tagging (TMT) proteomics. Eighteen lambs were allocated to two dietary treatment groups: control diet or control diet with the addition of 1% L-arginine. The results revealed that dietary arginine supplementation increased muscle fibre diameter and cross-sectional area (P < 0.05), which was attributable to protein deposition, as evidenced by increased RNA content, RNA/DNA ratio, inhibition of apoptotic enzyme activity, and alterations in the IGF-1/Akt signaling pathway (P < 0.05). In addition, dietary arginine elevated pH24h, a* values, and IMF content, decreased shear force value and backfat thickness (P < 0.05), as well as decreased the formation of lipid oxidation products involved in meat flavor including hexanal, heptanal, octanal, nonanal and 1-octen-3-ol by increasing the antioxidant capacity of the muscle (P < 0.05). The proteomics results suggested that seven enrichment pathways may be potential mechanisms by which arginine affected the muscle structure and meat characteristics of lambs. In summary, arginine supplementation in lamb diets provides a safe and effective way to improve meat quality, and antioxidant capacity of muscle of lamb.
Collapse
Affiliation(s)
- Lu Dou
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Chang Liu
- Inner Mongolia Vocational College of Chemical Engineering, Hohhot 010018, China
| | - Rina Su
- Inner Mongolia Vocational College of Chemical Engineering, Hohhot 010018, China
| | - Mirco Corazzin
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, 33100, Udine, Italy
| | - Zhimin Jin
- Inner Mongolia Autonomous Region Administration of Market Supervision Evaluation & Inspection Center, Hohhot 010018, China
| | - Zhihao Yang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Guanhua Hu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Min Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Lina Sun
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Lihua Zhao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Ye Jin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China
| | - Lin Su
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the people's Republic of China, Hohhot 010018, China.
| |
Collapse
|
7
|
Bakinowska E, Kiełbowski K, Pawlik A. The Role of MicroRNA in the Pathogenesis of Acute Kidney Injury. Cells 2024; 13:1559. [PMID: 39329743 PMCID: PMC11444149 DOI: 10.3390/cells13181559] [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: 06/26/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Acute kidney injury (AKI) describes a condition associated with elevated serum creatinine levels and decreased glomerular filtration rate. AKI can develop as a result of sepsis, the nephrotoxic properties of several drugs, and ischemia/reperfusion injury. Renal damage can be associated with metabolic acidosis, fluid overload, and ionic disorders. As the molecular background of the pathogenesis of AKI is insufficiently understood, more studies are needed to identify the key signaling pathways and molecules involved in the progression of AKI. Consequently, future treatment methods may be able to restore organ function more rapidly and prevent progression to chronic kidney disease. MicroRNAs (miRNAs) are small molecules that belong to the non-coding RNA family. Recently, numerous studies have demonstrated the altered expression profile of miRNAs in various diseases, including inflammatory and neoplastic conditions. As miRNAs are major regulators of gene expression, their dysregulation is associated with impaired homeostasis and cellular behavior. The aim of this article is to discuss current evidence on the involvement of miRNAs in the pathogenesis of AKI.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
8
|
Bérubé M, Abedini A, Lapointe E, Gusscott S, Brind'Amour J, Zamberlam G, Boerboom D. SFRP4 promotes autophagy and blunts FSH responsiveness through inhibition of AKT signaling in ovarian granulosa cells. Cell Commun Signal 2024; 22:396. [PMID: 39138534 PMCID: PMC11323480 DOI: 10.1186/s12964-024-01736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Secreted frizzled-related proteins (SFRPs) comprise a family of WNT signaling antagonists whose roles in the ovary are poorly understood. Sfrp4-null mice were previously found to be hyperfertile due to an enhanced granulosa cell response to gonadotropins, leading to decreased antral follicle atresia and enhanced ovulation rates. The present study aimed to elucidate the mechanisms whereby SFRP4 antagonizes FSH action. METHODS Primary cultures of granulosa cells from wild-type mice were treated with FSH and/or SFRP4, and effects of treatment on gene expression were evaluated by RT-qPCR and RNAseq. Bioinformatic analyses were conducted to analyse the effects of SFRP4 on the transcriptome, and compare them to those of FSH or a constitutively active mutant of FOXO1. Additional granulosa cell cultures from wild-type or Sfrp4-null mice, some pretreated with pharmacologic inhibitors of specific signaling effectors, were used to examine the effects of FSH and/or SFRP4 on signaling pathways, autophagy and apoptosis by western blotting and TUNEL. RESULTS Treatment of cultured granulosa cells with recombinant SFRP4 was found to decrease basal and FSH-stimulated mRNA levels of FSH target genes. Unexpectedly, this effect was found to occur neither via a canonical (CTNNB1-dependent) nor non-canonical WNT signaling mechanism, but was found to be GSK3β-dependent. Rather, SFRP4 was found to antognize AKT activity via a mechanism involving AMPK. This lead to the hypophosphorylation of FOXO1 and a decrease in the expression of a portion of the FSH and FOXO1 transcriptomes. Conversely, FSH-stimulated AMPK, AKT and FOXO1 phosphorylation levels were found to be increased in the granulosa cells of Sfrp4-null mice relative to wild-type controls. SFRP4 treatement of granulosa cells also induced autophagy by signaling via AKT-mTORC1-ULK1, as well as apoptosis. CONCLUSIONS This study identifies a novel GSK3β-AMPK-AKT signaling mechanism through which SFPR4 antagonizes FSH action, and further identifies SFRP4 as a novel regulator of granulosa cell autophagy. These findings provide a mechanistic basis for the phenotypic changes previously observed in Sfrp4-null mice, and broaden our understanding of the physiological roles of WNT signaling processes in the ovary.
Collapse
Affiliation(s)
- Michael Bérubé
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Atefeh Abedini
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Evelyne Lapointe
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Samuel Gusscott
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Julie Brind'Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada.
| |
Collapse
|
9
|
Culver SA, Hargett SR, Balugo JLLQ, Gildea JJ, Harris TE, Siragy HM. Nephron specific ATP6AP2 knockout increases urinary excretion of fatty acids and decreases renal cortical megalin expression. Sci Rep 2024; 14:18724. [PMID: 39134597 PMCID: PMC11319469 DOI: 10.1038/s41598-024-69749-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
ATP6AP2 knockout in the renal nephron impairs receptor-mediated endocytosis, increasing urinary albumin and glucose excretion and impairing weight gain. Nonesterified fatty acids (NEFA) in urine are bound to albumin and reabsorbed in the proximal tubule through receptor-mediated endocytosis by the megalin-cubilin complex. We hypothesized that ATP6AP2 knockout increases urinary NEFA excretion through a reduction in megalin. Ten-week-old male C57BL/6 mice with nephron specific inducible ATP6AP2 knockout and noninduced controls were fed either normal diet (ND 12% fat) or high fat diet (HFD 45% fat) for 6 months. ATP6AP2 knockout significantly increased urine albumin:creatinine ratio in both ND and HFD fed mice while normalized urine NEFA concentration increased 489% and 259% in ND and HFD knockout mice compared to respective controls. Knockout decreased renal cortical megalin mRNA by 47% on ND and 49% on HFD while megalin protein expression decreased by 36% and 44% respectively. At the same time, markers of mTOR activity were increased while autophagy was impaired. Our results indicate that nephron specific ATP6AP2 knockout increases urinary NEFA excretion in the setting of impaired receptor-mediated endocytosis. Further investigation should determine whether ATP6AP2 contributes to obesity related ectopic lipid deposition in the proximal tubule.
Collapse
Affiliation(s)
- Silas A Culver
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, P.O. Box 801409, Charlottesville, VA, 22908-1409, USA.
| | - Stefan R Hargett
- Department of Pharmacology, University of Virginia Health System, Charlottesville, USA
| | - Jamie L L Q Balugo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, P.O. Box 801409, Charlottesville, VA, 22908-1409, USA
| | - John J Gildea
- Department of Pathology, University of Virginia Health System, Charlottesville, USA
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia Health System, Charlottesville, USA
| | - Helmy M Siragy
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, P.O. Box 801409, Charlottesville, VA, 22908-1409, USA
| |
Collapse
|
10
|
Bao Y, Shan Q, Lu K, Yang Q, Liang Y, Kuang H, Wang L, Hao M, Peng M, Zhang S, Cao G. Renal tubular epithelial cell quality control mechanisms as therapeutic targets in renal fibrosis. J Pharm Anal 2024; 14:100933. [PMID: 39247486 PMCID: PMC11377145 DOI: 10.1016/j.jpha.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 09/10/2024] Open
Abstract
Renal fibrosis is a devastating consequence of progressive chronic kidney disease, representing a major public health challenge worldwide. The underlying mechanisms in the pathogenesis of renal fibrosis remain unclear, and effective treatments are still lacking. Renal tubular epithelial cells (RTECs) maintain kidney function, and their dysfunction has emerged as a critical contributor to renal fibrosis. Cellular quality control comprises several components, including telomere homeostasis, ubiquitin-proteasome system (UPS), autophagy, mitochondrial homeostasis (mitophagy and mitochondrial metabolism), endoplasmic reticulum (ER, unfolded protein response), and lysosomes. Failures in the cellular quality control of RTECs, including DNA, protein, and organelle damage, exert profibrotic functions by leading to senescence, defective autophagy, ER stress, mitochondrial and lysosomal dysfunction, apoptosis, fibroblast activation, and immune cell recruitment. In this review, we summarize recent advances in understanding the role of quality control components and intercellular crosstalk networks in RTECs, within the context of renal fibrosis.
Collapse
Affiliation(s)
- Yini Bao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiyuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ying Liang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haodan Kuang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shuosheng Zhang
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030600, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310009, China
| |
Collapse
|
11
|
Preechanukul A, Saiprom N, Rochaikun K, Moonmueangsan B, Phunpang R, Ottiwet O, Kongphrai Y, Wapee S, Janon R, Dunachie S, Kronsteiner B, Chantratita N. Metabolic requirements of CD160 expressing memory-like NK cells in Gram-negative bacterial infection. Clin Transl Immunology 2024; 13:e1513. [PMID: 38957437 PMCID: PMC11218174 DOI: 10.1002/cti2.1513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 07/04/2024] Open
Abstract
Objective Unique metabolic requirements accompany the development and functional fates of immune cells. How cellular metabolism is important in natural killer (NK) cells and their memory-like differentiation in bacterial infections remains elusive. Methods Here, we utilise our established NK cell memory assay to investigate the metabolic requirement for memory-like NK cell formation and function in response to the Gram-negative intracellular bacteria Burkholderia pseudomallei (BP), the causative agent of melioidosis. Results We demonstrate that CD160+ memory-like NK cells upon BP stimulation upregulate glucose and amino acid transporters in a cohort of recovered melioidosis patients which is maintained at least 3-month post-hospital admission. Using an in vitro assay, human BP-specific CD160+ memory-like NK cells show metabolic priming including increased expression of glucose and amino acid transporters with elevated glucose uptake, increased mTOR activation and mitochondrial membrane potential upon BP re-stimulation. Antigen-specific and cytokine-induced IFN-γ production of this memory-like NK cell subset are highly dependent on oxidative phosphorylation (OXPHOS) with some dependency on glycolysis, whereas the formation of CD160+ memory-like NK cells in vitro is dependent on fatty acid oxidation and OXPHOS and further increased by metformin. Conclusion This study reveals the link between metabolism and cellular function of memory-like NK cells, which can be exploited for vaccine design and for monitoring protection against Gram-negative bacterial infection.
Collapse
Affiliation(s)
- Anucha Preechanukul
- Department of Microbiology and Immunology, Faculty of Tropical Medicine Mahidol University Bangkok Thailand
- Division of Infection and Immunity University College London London UK
| | - Natnaree Saiprom
- Department of Microbiology and Immunology, Faculty of Tropical Medicine Mahidol University Bangkok Thailand
| | - Kitilak Rochaikun
- Department of Microbiology and Immunology, Faculty of Tropical Medicine Mahidol University Bangkok Thailand
| | - Boonthanom Moonmueangsan
- Department of Microbiology and Immunology, Faculty of Tropical Medicine Mahidol University Bangkok Thailand
| | - Rungnapa Phunpang
- Department of Microbiology and Immunology, Faculty of Tropical Medicine Mahidol University Bangkok Thailand
| | - Orawan Ottiwet
- Department of Medical Technology and Clinical Pathology Mukdahan Hospital Mukdahan Thailand
| | - Yuphin Kongphrai
- Department of Medical Technology and Clinical Pathology Mukdahan Hospital Mukdahan Thailand
| | - Soonthon Wapee
- Department of Medical Technology and Clinical Pathology Mukdahan Hospital Mukdahan Thailand
| | - Rachan Janon
- Department of Medicine Mukdahan Hospital Mukdahan Thailand
| | - Susanna Dunachie
- Mahidol-Oxford Tropical Medicine Research Unit Mahidol University Bangkok Thailand
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine University of Oxford Oxford UK
- Nuffield Department of Clinical Medicine, NDM Centre for Global Health Research University of Oxford Oxford UK
- Oxford University Hospitals NHS Foundation Trust Oxford UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine University of Oxford Oxford UK
- Nuffield Department of Clinical Medicine, NDM Centre for Global Health Research University of Oxford Oxford UK
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine Mahidol University Bangkok Thailand
- Mahidol-Oxford Tropical Medicine Research Unit Mahidol University Bangkok Thailand
| |
Collapse
|
12
|
Katerelos M, Gleich K, Harley G, Loh K, Oakhill JS, Kemp BE, de Souza DP, Narayana VK, Coughlan MT, Laskowski A, Ling NXY, Murray-Segal L, Brink R, Lee M, Power DA, Mount PF. The AMPK activator ATX-304 alters cellular metabolism to protect against cisplatin-induced acute kidney injury. Biomed Pharmacother 2024; 175:116730. [PMID: 38749175 DOI: 10.1016/j.biopha.2024.116730] [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: 01/05/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024] Open
Abstract
Acute kidney injury (AKI) disrupts energy metabolism. Targeting metabolism through AMP-activated protein kinase (AMPK) may alleviate AKI. ATX-304, a pan-AMPK activator, was evaluated in C57Bl/6 mice and tubular epithelial cell (TEC) cultures. Mice received ATX-304 (1 mg/g) or control chow for 7 days before cisplatin-induced AKI (CI-AKI). Primary cultures of tubular epithelial cells (TECs) were pre-treated with ATX-304 (20 µM, 4 h) prior to exposure to cisplatin (20 µM, 23 h). ATX-304 increased acetyl-CoA carboxylase phosphorylation, indicating AMPK activation. It protected against CI-AKI measured by serum creatinine (control 0.05 + 0.03 mM vs ATX-304 0.02 + 0.01 mM, P = 0.03), western blot for neutrophil gelatinase-associated lipocalin (NGAL) (control 3.3 + 1.8-fold vs ATX-304 1.2 + 0.55-fold, P = 0.002), and histological injury (control 3.5 + 0.59 vs ATX-304 2.7 + 0.74, P = 0.03). In TECs, pre-treatment with ATX-304 protected against cisplatin-mediated injury, as measured by lactate dehydrogenase release, MTS cell viability, and cleaved caspase 3 expression. ATX-304 protection against cisplatin was lost in AMPK-null murine embryonic fibroblasts. Metabolomic analysis in TECs revealed that ATX-304 (20 µM, 4 h) altered 66/126 metabolites, including fatty acids, tricarboxylic acid cycle metabolites, and amino acids. Metabolic studies of live cells using the XFe96 Seahorse analyzer revealed that ATX-304 increased the basal TEC oxygen consumption rate by 38%, whereas maximal respiration was unchanged. Thus, ATX-304 protects against cisplatin-mediated kidney injury via AMPK-dependent metabolic reprogramming, revealing a promising therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Marina Katerelos
- Department of Nephrology, Austin Health, Heidelberg, Victoria 3084, Australia; Kidney Laboratory, The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria 3084, Australia
| | - Kurt Gleich
- Department of Nephrology, Austin Health, Heidelberg, Victoria 3084, Australia; Kidney Laboratory, The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria 3084, Australia
| | - Geoff Harley
- Department of Nephrology, Austin Health, Heidelberg, Victoria 3084, Australia; Kidney Laboratory, The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria 3084, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - David P de Souza
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Vinod K Narayana
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Melinda T Coughlan
- Glycation, Nutrition and Metabolism Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Parkville, Victoria 3052, Australia
| | - Adrienne Laskowski
- Glycation, Nutrition and Metabolism Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Naomi X Y Ling
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Lisa Murray-Segal
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia; St. Vincent's Clinical School, University of New South Wales, St. Vincent's Hospital, Darlinghurst, New South Wales 2010, Australia
| | - Mardiana Lee
- Department of Nephrology, Austin Health, Heidelberg, Victoria 3084, Australia; Kidney Laboratory, The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria 3084, Australia
| | - David A Power
- Department of Nephrology, Austin Health, Heidelberg, Victoria 3084, Australia; Kidney Laboratory, The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria 3084, Australia; Department of Medicine (Austin), The University of Melbourne, Heidelberg ,Victoria 3084, Australia
| | - Peter F Mount
- Department of Nephrology, Austin Health, Heidelberg, Victoria 3084, Australia; Kidney Laboratory, The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria 3084, Australia; Department of Medicine (Austin), The University of Melbourne, Heidelberg ,Victoria 3084, Australia.
| |
Collapse
|
13
|
Peng M, Ye F, Fan C, Dong J, Chai W, Deng W, Zhang H, Yang L. Endogenous S100P-mediated autophagy regulates the chemosensitivity of leukemia cells through the p53/AMPK/mTOR pathway. Am J Cancer Res 2024; 14:1121-1138. [PMID: 38590396 PMCID: PMC10998763 DOI: 10.62347/nwxe8730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 03/02/2024] [Indexed: 04/10/2024] Open
Abstract
Autophagy, a highly regulated lysosome-dependent catabolic pathway, has garnered increasing attention because of its role in leukemia resistance. Among the S100 family of small calcium-binding proteins, S100P is differentially expressed in various tumor cell lines, thereby influencing tumor occurrence, invasion, metastasis, and drug resistance. However, the relationship between S100P and autophagy in determining chemosensitivity in leukemia cells remains unexplored. Our investigation revealed a negative correlation between S100P expression and the clinical status in childhood leukemia, with its presence observed in HL-60 and Jurkat cell lines. Suppression of S100P expression resulted in increased cell proliferation and decreased chemosensitivity in leukemia cells, whereas enhancement of S100P expression inhibited cell proliferation and increased chemosensitivity. Additionally, S100P knockdown drastically promoted autophagy, which was subsequently suppressed by S100P upregulation. Moreover, the p53/AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway was found to be functionally associated with S100P-mediated autophagy. Knockdown of S100P expression led to a decrease in p53 and p-mTOR levels and an increase in p-AMPK expression, ultimately promoting autophagy. This effect was reversed by administration of Tenovin-6 (a p53 activator) and Compound C (an AMPK inhibitor). The findings of our in vivo experiments provide additional evidence supporting the aforementioned data. Specifically, S100P inhibition significantly enhanced the growth of HL-60 tumor xenografts and increased the expression of microtubule-associated protein 1 light chain 3 and p-AMPK in nude mice. Consequently, it can be concluded that S100P plays a regulatory role in the chemosensitivity of leukemia cells by modulating the p53/AMPK/mTOR pathway, which controls autophagy in leukemia cells.
Collapse
Affiliation(s)
- Min Peng
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Fanghua Ye
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Chenying Fan
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Jiajia Dong
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Wenwen Chai
- Department of Nuclear Medicine, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Wenjun Deng
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Hui Zhang
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Liangchun Yang
- Department of Pediatrics, Xiangya Hospital Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| |
Collapse
|
14
|
Yuan P, Feng A, Wei Y, Li S, Fu Y, Wang X, Guo M, Feng W, Zheng X. Indole-3-carboxaldehyde alleviates cisplatin-induced acute kidney injury in mice by improving mitochondrial dysfunction via PKA activation. Food Chem Toxicol 2024; 186:114546. [PMID: 38408633 DOI: 10.1016/j.fct.2024.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Cisplatin (DDP) is widely used in the treatment of cancer as a chemotherapeutic drug. However, its severe nephrotoxicity limits the extensive application of cisplatin, which is characterized by injury and apoptosis of renal tubular epithelial cells. This study aimed to reveal the protective effect and its underlying mechanism of Indole-3-carboxaldehyde (IC) against DDP-induced AKI in mice and NRK-52E cells pretreated with PKA antagonist (H-89). Here, we reported that IC improved renal artery blood flow velocity and renal function related indicators, attenuated renal pathological changes, which were confirmed by the results of HE staining and PASM staining. Meanwhile, IC inhibited the levels of inflammatory factors, oxidative stress, CTR1, OCT2, and the levels of autophagy and apoptosis. Mitochondrial dysfunction was significantly improved as observed by TEM. To clarify the potential mechanism, NRK-52E cells induced by DDP was used and the results proved that H-89 could blocked the improvement with IC effectively in vitro. Our findings showed that IC has the potential to treat cisplatin-induced AKI, and its role in protecting the kidney was closely related to activating PKA, inhibiting autophagy and apoptosis, improving mitochondrial function, which could provide a theoretical basis for the development of new clinical drugs.
Collapse
Affiliation(s)
- Peipei Yuan
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China
| | - Aozi Feng
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Yaxin Wei
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Saifei Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yang Fu
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Xiao Wang
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Menghuan Guo
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Weisheng Feng
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, PR China.
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, 450046, PR China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, PR China
| |
Collapse
|
15
|
Gao J, Deng Q, Yu J, Wang C, Wei W. Role of renal tubular epithelial cells and macrophages in cisplatin-induced acute renal injury. Life Sci 2024; 339:122450. [PMID: 38262575 DOI: 10.1016/j.lfs.2024.122450] [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: 08/01/2023] [Revised: 12/30/2023] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a sudden and continuous decline in renal function. The drug cisplatin is commonly used as chemotherapy for solid tumors, and cisplatin-induced acute kidney injury (CI-AKI), which is characterized by acute tubular necrosis and inflammation, frequently occurs in tumor patients. Renal tubular epithelial cells (RTECs) are severely damaged early in this process and play an important role in renal tubular injury and the recruitment of immune cells. Macrophages are the most common infiltrating immune cells in the kidney and have a significant impact on CI-AKI and subsequent repair. This article reviews the latest research progress on the effects of RTECs and macrophages on CI-AKI and their interactions in AKI to provide a direction for identifying therapeutic targets for treating AKI.
Collapse
Affiliation(s)
- Jinzhang Gao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, China; Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| | - Qinxiang Deng
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Third Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Jun Yu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, China; Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, China; Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, China; Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China.
| |
Collapse
|
16
|
Gao X, Wu Y. Perioperative acute kidney injury: The renoprotective effect and mechanism of dexmedetomidine. Biochem Biophys Res Commun 2024; 695:149402. [PMID: 38159412 DOI: 10.1016/j.bbrc.2023.149402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Dexmedetomidine (DEX) is a highly selective and potent α2-adrenoceptor (α2-AR) agonist that is widely used as a clinical anesthetic to induce anxiolytic, sedative, and analgesic effects. In recent years, a growing body of evidence has demonstrated that DEX protects against acute kidney injury (AKI) caused by sepsis, drugs, surgery, and ischemia-reperfusion (I/R) in organs or tissues, indicating its potential role in the prevention and treatment of AKI. In this review, we summarized the evidence of the renoprotective effects of DEX on different models of AKI and explored the mechanism. We found that the renoprotective effects of DEX mainly involved antisympathetic effects, reducing inflammatory reactions and oxidative stress, reducing apoptosis, increasing autophagy, reducing ferroptosis, protecting renal tubular epithelial cells (RTECs), and inhibiting renal fibrosis. Thus, the use of DEX is a promising strategy for the management and treatment of perioperative AKI. The aim of this review is to further clarify the renoprotective mechanism of DEX to provide a theoretical basis for its use in basic research in various AKI models, clinical management, and the treatment of perioperative AKI.
Collapse
Affiliation(s)
- Xiong Gao
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yaohua Wu
- Department of Anesthesiology, Huanggang Central Hospital, Huanggang, Hube, China.
| |
Collapse
|
17
|
Luo D, Li S, Guo J, Yue H, Shi L, Liu R, Wang J, Shi X. The role and mechanism of AZD5363 anti-leukemia activity in T-cell acute lymphoblastic leukemia. Eur J Pharmacol 2024; 963:176268. [PMID: 38096965 DOI: 10.1016/j.ejphar.2023.176268] [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: 07/11/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive and heterogeneous hematologic malignancy. Chemotherapy resistance and refractory relapses are the most important challenges in T-ALL. PI3K/Akt/mTOR pathway has been implicated in regulating cell survival, T-ALL development and resistance to chemotherapy. We explored the effects of AZD5363 (a potent pan-Akt inhibitor) alone and in combination with autophagy inhibitor hydroxycholoroquine sulfate (HCQ) in cultured CCRF-CEM, Jurkat and PF382 cells and a T-ALL xenograft mouse model. METHODS A xenograft mouse model was used to investigate the effect of AZD5363 on T-ALL progression. MTT assay, flow cytometry, siRNA, transmission electron microscopy and western blotting were performed in cultured CCRF-CEM, Jurkat and PF382 cells. The interaction between AZD5363 and HCQ was explored by molecular docking. RESULTS AZD5363 delayed T-ALL progression and increased the expression of cleaved caspase-3 and LC3B-II in mice. AZD5363 decreased cells viability by arresting cell cycle in the G1 phase and inducing apoptosis, and, significantly increased the number of autophagosomes (p < 0.01). The increased expression of cleaved caspase-3 and LC3B-II, and phosphorylation of Akt and mTOR were significantly, inhibited by AZD5363. HCQ blocked AZD5363-induced autophagy and enhanced AZD5363-induced cell death (p < 0.01). CONCLUSIONS AZD5363 suppressed T-ALL progression and its anti-leukemia activity was enhanced by HCQ in T-ALL cells, which might provide a potential therapeutic strategy for human T-ALL.
Collapse
Affiliation(s)
- Danqing Luo
- Department of Hematology, The Affiliated of Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shen Li
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Jin Guo
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Huixuan Yue
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lixiao Shi
- Department of Hematology, The Affiliated of Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Rong Liu
- Department of Hematology, The Affiliated of Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Jianhua Wang
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Xiaodong Shi
- Department of Hematology, The Affiliated of Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
18
|
Anitha K, Chenchula S, Shama N, Mishra N, Singh MK, Radhika C. Molecular Mechanisms of Autophagy Regulation in Cancer. CANCER DRUG DISCOVERY AND DEVELOPMENT 2024:73-93. [DOI: 10.1007/978-3-031-66421-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Wang J, Zhou L, Hou H, Li J, Zhao X, Li J, Li J, Niu X, Hou R, Zhang K. IL-17A is involved in the hyperplasia of blood vessels in local lesions of psoriasis by inhibiting autophagy. J Cosmet Dermatol 2024; 23:326-338. [PMID: 37635345 DOI: 10.1111/jocd.15975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE Increased angiogenesis is a pathological feature of psoriasis, but the pathomechanisms of angiogenesis in psoriasis are not clear. Interleukin-17A (IL-17A) is the major effect factor in the pathogenesis of psoriasis. Our results showed that IL-17A can promote angiogenesis and cause endothelial cell inflammation. Autophagy plays an important role not only in regulating inflammation, but also in regulating angiogenesis. Whether angiogenesis in psoriasis is related to autophagy remains unclear. In this study, we treated human umbilical vein endothelial cells (HUVECs) with IL-17A to simulate increased angiogenesis to study whether increased angiogenesis in psoriasis is related to autophagy. METHODS AND RESULTS Our results showed that treatment of HUVECs with IL-17A significantly increased angiogenesis and expression levels of mRNA for multiple proinflammatory cytokines (CCL20, IL-8, CCL2, IL-6, and IL-1β) and, while decreasing intracellular levels of nitric oxide (NO) and NO synthase (NOS) activity. Moreover, IL-17A inhibited autophagy as shown that IL-17A significantly increased expression levels of LC3II and p62 proteins. Induction of autophagy ameliorated IL-17A-mediated inflammatory response and inhibited angiogenesis, accompanied by increased p-AMPKα(Thr172) and p-ULK1(Ser555) expression, and decreased p-mTOR(Ser2448) and p-ULK1(Ser757) expression. Furthermore, inhibition of either AMPK or lysosomal acidification completely overrode autophagy-induced changes in angiogenesis and NOS activity. Finally, induction of autophagy decreased apoptosis and caspase-3 activity in IL-17A-treated HUVECs. CONCLUSIONS These results showed that IL-17A is involved in angiogenesis and inflammatory response by inhibiting autophagy through AMPK signaling pathway, suggesting that autophagy may be a new therapeutic target for psoriasis.
Collapse
Affiliation(s)
- Juanjuan Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Ling Zhou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Hui Hou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiao Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Xincheng Zhao
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiajie Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuping Niu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruixia Hou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| |
Collapse
|
20
|
Owaki R, Aoki H, Toriuchi K, Inoue Y, Hayashi H, Takeshita S, Kakita H, Yamada Y, Aoyama M. AMPK activators suppress cholesterol accumulation in macrophages via suppression of the mTOR pathway. Exp Cell Res 2023; 432:113784. [PMID: 37730144 DOI: 10.1016/j.yexcr.2023.113784] [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: 05/11/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Atherosclerosis is a persistent inflammatory state that contributes significantly to cardiovascular disease, a primary cause of mortality worldwide. Enhanced lipid uptake by macrophages and their transformation into foam cells play a key role in the development of atherosclerosis. Recent studies using in vivo mouse models indicated that activation of AMPK has anti-atherosclerotic effects by upregulating the expression of cholesterol efflux transporters in foam cells and promoting cholesterol efflux. However, the pathway downstream of AMPK that contributes to elevated expression of cholesterol efflux transporters remains unclear. In this study, we found that activation of AMPK by AICAR and metformin inhibits foam cell formation via suppression of mTOR in macrophages. Specifically, activation of AMPK indirectly reduced the phosphorylation level of mTOR at Ser2448 and promoted the expression of cholesterol efflux transporters and cholesterol efflux. These inhibitory effects on foam cell formation were counteracted by mTOR activators. Metformin, a more nonspecific AMPK activator than AICAR, appears to inhibit foam cell formation via anti-inflammatory effects in addition to suppression of the mTOR pathway. The results of this study suggest that the development of new drugs targeting AMPK activation and mTOR inhibition may lead to beneficial results in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Reina Owaki
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Kohki Toriuchi
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Yasumichi Inoue
- Department of Cell Signaling, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Satoru Takeshita
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan; Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Hiroki Kakita
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan; Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Yasumasa Yamada
- Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
21
|
Jayaraman P, Rajagopal M, Paranjpe I, Liharska L, Suarez-Farinas M, Thompson R, Del Valle DM, Beckmann N, Oh W, Gulamali FF, Kauffman J, Gonzalez-Kozlova E, Dellepiane S, Vasquez-Rios G, Vaid A, Jiang J, Chen A, Sakhuja A, Chen S, Kenigsberg E, He JC, Coca SG, Chan L, Schadt E, Merad M, Kim-Schulze S, Gnjatic S, Tsalik E, Langley R, Charney AW, Nadkarni GN. Peripheral Transcriptomics in Acute and Long-Term Kidney Dysfunction in SARS-CoV2 Infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.25.23297469. [PMID: 37961671 PMCID: PMC10635190 DOI: 10.1101/2023.10.25.23297469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Acute kidney injury (AKI) is common in hospitalized patients with SARS-CoV2 infection despite vaccination and leads to long-term kidney dysfunction. However, peripheral blood molecular signatures in AKI from COVID-19 and their association with long-term kidney dysfunction are yet unexplored. Methods In patients hospitalized with SARS-CoV2, we performed bulk RNA sequencing using peripheral blood mononuclear cells(PBMCs). We applied linear models accounting for technical and biological variability on RNA-Seq data accounting for false discovery rate (FDR) and compared functional enrichment and pathway results to a historical sepsis-AKI cohort. Finally, we evaluated the association of these signatures with long-term trends in kidney function. Results Of 283 patients, 106 had AKI. After adjustment for sex, age, mechanical ventilation, and chronic kidney disease (CKD), we identified 2635 significant differential gene expressions at FDR<0.05. Top canonical pathways were EIF2 signaling, oxidative phosphorylation, mTOR signaling, and Th17 signaling, indicating mitochondrial dysfunction and endoplasmic reticulum (ER) stress. Comparison with sepsis associated AKI showed considerable overlap of key pathways (48.14%). Using follow-up estimated glomerular filtration rate (eGFR) measurements from 115 patients, we identified 164/2635 (6.2%) of the significantly differentiated genes associated with overall decrease in long-term kidney function. The strongest associations were 'autophagy', 'renal impairment via fibrosis', and 'cardiac structure and function'. Conclusions We show that AKI in SARS-CoV2 is a multifactorial process with mitochondrial dysfunction driven by ER stress whereas long-term kidney function decline is associated with cardiac structure and function and immune dysregulation. Functional overlap with sepsis-AKI also highlights common signatures, indicating generalizability in therapeutic approaches. SIGNIFICANCE STATEMENT Peripheral transcriptomic findings in acute and long-term kidney dysfunction after hospitalization for SARS-CoV2 infection are unclear. We evaluated peripheral blood molecular signatures in AKI from COVID-19 (COVID-AKI) and their association with long-term kidney dysfunction using the largest hospitalized cohort with transcriptomic data. Analysis of 283 hospitalized patients of whom 37% had AKI, highlighted the contribution of mitochondrial dysfunction driven by endoplasmic reticulum stress in the acute stages. Subsequently, long-term kidney function decline exhibits significant associations with markers of cardiac structure and function and immune mediated dysregulation. There were similar biomolecular signatures in other inflammatory states, such as sepsis. This enhances the potential for repurposing and generalizability in therapeutic approaches.
Collapse
|
22
|
Bu X, Zhang J, Sun H, Wang W, Ma X, Sun L. SEC61 translocon subunit gamma enhances low-dose cisplatin-induced cancer-stem cell properties of head and neck squamous cell carcinoma via enhancing Ca 2+-mediated autophagy. J Dent Sci 2023; 18:1685-1692. [PMID: 37799908 PMCID: PMC10547946 DOI: 10.1016/j.jds.2023.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
Background/purpose High SEC61 translocon subunit gamma (SEC61G) expression is associated with an unfavorable prognosis in patients with head and neck squamous cell carcinoma (HNSCC), but the underlying mechanisms remain poorly understood. Materials and methods HNSCC representative cell lines SCC15 and CAL27 were used to explore the regulation of SEC61G on Ca2+ leak from the endoplasmic reticulum (ER). Ca2+-activated autophagy was monitored by fluorescent labeling of autophagosomes and western blotting assays. CSC marker expression, sphere formation, colony formation, and transwell of invasion were detected to investigate the role of SEC61G in regulating cancer-stem cell (CSC) properties. Results Among the SEC61 complex genes, only SEC61G upregulation is consistently associated with unfavorable progression-free interval and disease-specific survival in patients with HNSCC. Low-dose cisplatin (CDDP) treatment induced SEC61G upregulation in SCC15 and CAL27 cells. SEC61G knockdown significantly impaired CDDP-induced Ca2+ from the ER and the phosphorylation of ERK1/2 and AMPK. CDDP-induced autophagy in HNSCC cells were hampered by SEC61G shRNA, in terms of impaired autophagosome formation, lowered LC3-II/GAPDH ratio and restored p62 expression. CDDP-induced CSC properties, including CSC marker expression, sphere formation, colony formation, and invasive capabilities could be suppressed by shSEC61G and chloroquine, a specific autophagy inhibitor. Conclusion Findings of this study revealed the contribution of SEC61G in promoting cisplatin-induced CSC properties of HNSCC cells via promoting Ca2+-mediated autophagy.
Collapse
Affiliation(s)
- Xiangbin Bu
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jun Zhang
- Department of Interventional Catheter Room, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Hengyan Sun
- Department of Stomatology, Yantai Penglai Traditional Chinese Medicine Hospital, Shandong, China
| | - Wenlong Wang
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiangrui Ma
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Legang Sun
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| |
Collapse
|
23
|
Wang RL, Liu SH, Shen SH, Jian LY, Yuan Q, Guo HH, Huang JS, Chen PH, Huang RF. Protective Mechanism of Cordyceps sinensis Treatment on Acute Kidney Injury-Induced Acute Lung Injury through AMPK/mTOR Signaling Pathway. Chin J Integr Med 2023; 29:875-884. [PMID: 36843056 DOI: 10.1007/s11655-023-3593-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2022] [Indexed: 02/28/2023]
Abstract
OBJECTIVE To investigate protective effect of Cordyceps sinensis (CS) through autophagy-associated adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway in acute kidney injury (AKI)-induced acute lung injury (ALI). METHODS Forty-eight male Sprague-Dawley rats were divided into 4 groups according to a random number table, including the normal saline (NS)-treated sham group (sham group), NS-treated ischemia reperfusion injury (IRI) group (IRI group), and low- (5 g/kg·d) and high-dose (10 g/kg·d) CS-treated IRI groups (CS1 and CS2 groups), 12 rats in each group. Nephrectomy of the right kidney was performed on the IRI rat model that was subjected to 60 min of left renal pedicle occlusion followed by 12, 24, 48, and 72 h of reperfusion. The wet-to-dry (W/D) ratio of lung, levels of serum creatinine (Scr), blood urea nitrogen (BUN), inflammatory cytokines such as interleukin- β and tumor necrosis factor- α, and biomarkers of oxidative stress such as superoxide dismutase, malonaldehyde (MDA) and myeloperoxidase (MPO), were assayed. Histological examinations were conducted to determine damage of tissues in the kidney and lung. The protein expressions of light chain 3 II/light chain 3 I (LC3-II/LC3-I), uncoordinated-51-like kinase 1 (ULK1), P62, AMPK and mTOR were measured by Western blot and immunohistochemistry, respectively. RESULTS The renal IRI induced pulmonary injury following AKI, resulting in significant increases in W/D ratio of lung, and the levels of Scr, BUN, inflammatory cytokines, MDA and MPO (P<0.01); all of these were reduced in the CS groups (P<0.05 or P<0.01). Compared with the IRI groups, the expression levels of P62 and mTOR were significantly lower (P<0.05 or P<0.01), while those of LC3-II/LC3-I, ULK1, and AMPK were significantly higher in the CS2 group (P<0.05 or P<0.01). CONCLUSION CS had a potential in treating lung injury following renal IRI through activation of the autophagy-related AMPK/mTOR signaling pathway in AKI-induced ALI.
Collapse
Affiliation(s)
- Ruo-Lin Wang
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shu-Hua Liu
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Si-Heng Shen
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lu-Yong Jian
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qi Yuan
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
- The Third Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Hua-Hui Guo
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
| | - Jia-Sheng Huang
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
| | - Peng-Hui Chen
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China
| | - Ren-Fa Huang
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, 518034, China.
| |
Collapse
|
24
|
Wang P, Chen W, Zhao S, Cheng F. The role of LncRNA-regulated autophagy in AKI. Biofactors 2023; 49:1010-1021. [PMID: 37458310 DOI: 10.1002/biof.1980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/16/2023] [Indexed: 10/04/2023]
Abstract
Acute kidney injury (AKI) is a complex clinical syndrome involving a series of pathophysiological processes regulated by multiple pathways at the molecular and cellular level. Long noncoding RNAs (lncRNAs) play an important role in the regulation of epigenetics, and their regulation of autophagy-related genes in AKI has attracted increasing attention. However, the role of lncRNA-regulated autophagy in AKI has not been fully elucidated. Evidence indicated that lncRNAs play regulatory roles in most factors that induce AKI. LncRNAs can regulate autophagy in AKI via a complex network of regulatory pathways to affect the development and prognosis of AKI. This article reviewed and analyzed the pathways of lncRNA regulation of autophagy in AKI in recent years. The results provide new ideas for further study of the pathophysiological process and targeted therapy for AKI.
Collapse
Affiliation(s)
- Peihan Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
25
|
Tang Y, Liu Y, Zhou H, Lu H, Zhang Y, Hua J, Liao X. Esketamine is neuroprotective against traumatic brain injury through its modulation of autophagy and oxidative stress via AMPK/mTOR-dependent TFEB nuclear translocation. Exp Neurol 2023; 366:114436. [PMID: 37187276 DOI: 10.1016/j.expneurol.2023.114436] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023]
Abstract
Recent clinical studies highlight the neuroprotective effects of esketamine, but its benefits following traumatic brain injury (TBI) have not been defined. Here, we investigated the effects of esketamine following TBI and its associated neuroprotection mechanisms. In our study, controlled cortical impact injury on mice was utilized to induce the TBI model in vivo. TBI mice were randomized to receive vehicle or esketamine at 2 h post-injury for 7 consecutive days. Neurological deficits and brain water content in mice were detected, respectively. Cortical tissues surrounding focal trauma were obtained for Nissl staining, immunofluorescence, immunohistochemistry, and ELISA assay. In vitro, esketamine were added in culture medium after cortical neuronal cells induced by H2O2 (100μM). After exposed for 12h, neuronal cells were obtained for western blotting, immunofluorescence, ELISA and CO-IP assay. Following administration of 2-8 mg/kg esketamine, we observed that 8 mg/kg esketamine produced no additional recovery of neurological function and ability to alleviate brain edema in TBI mice model, so 4 mg/kg esketamine was selected for subsequent experiments. Additionally, esketamine can effectively reduce TBI-induced oxidative stress, the number of damaged neurons, and the number of TUNEL-positive cells in the cortex of TBI models. Meanwhile, the levels of Beclin 1, LC3 II, and the number of LC3-positive cells in injured cortex were also increased following esketamine exposure. Western blotting and immunofluorescence assays showed that esketamine accelerated the nuclear translocation of TFEB, increased the p-AMPKα level and decreased the p-mTOR level. Similar results including nuclear translocation of TFEB, the increases of autophagy-related markers, and influences of AMPK/mTOR pathway were observed in H2O2-induced cortical neuronal cells; however, BML-275 (AMPK inhibitor) can reverse these effects of esketamine. Furthermore, TFEB silencing not only decreased the Nrf2 level in H2O2-induced cortical neuronal cells, but also alleviated the oxidative stress. Importantly, CO-IP confirmed the interaction between TFEB and Nrf2 in cortical neuronal cells. These findings suggested that esketamine exerts the neuroprotective effects of esketamine in TBI mice model via enhancing autophagy and alleviating oxidative stress; its mechanism involves AMPK/mTOR-dependent TFEB nuclear translocation-induced autophagy and TFEB/Nrf2-induced antioxidant system.
Collapse
Affiliation(s)
- Yanbin Tang
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China
| | - Yufang Liu
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China
| | - Huanzhu Zhou
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China; School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Haibo Lu
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China
| | - Yafei Zhang
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China
| | - Jun Hua
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China
| | - Xingzhi Liao
- Department of Anesthesiology, 904th Hospital of The Joint Logistics Support Force of the PLA, Wuxi 214044, Jiangsu, China.
| |
Collapse
|
26
|
Yu B, Jin L, Yao X, Zhang Y, Zhang G, Wang F, Su X, Fang Q, Xiao L, Yang Y, Jiang LH, Chen J, Yang W, Lin W, Han F. TRPM2 protects against cisplatin-induced acute kidney injury and mitochondrial dysfunction via modulating autophagy. Theranostics 2023; 13:4356-4375. [PMID: 37649595 PMCID: PMC10465213 DOI: 10.7150/thno.84655] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/26/2023] [Indexed: 09/01/2023] Open
Abstract
Background: Cisplatin is a widely used anti-tumor agent but its use is frequently limited by nephrotoxicity. Transient receptor potential melastatin 2 (TRPM2) is a non-selective cation channel which is generally viewed as a sensor of oxidative stress, and increasing evidence supports its link with autophagy, a critical process for organelle homeostasis. Methods: Cisplatin-induced cell injury and mitochondrial damage were both assessed in WT and Trpm2-knockout mice and primary cells. RNA sequencing, immunofluorescence staining, immunoblotting and flowcytometry were applied to interpret the mechanism of TRPM2 in cisplatin nephrotoxicity. Results: Knockout of TRPM2 exacerbates renal dysfunction, tubular injury and cell apoptosis in a model of acute kidney injury (AKI) induced by treatment with cisplatin. Cisplatin-caused tubular mitochondrial damage is aggravated in TRPM2-deficient mice and cells and, conversely, alleviated by treatment with Mito-TEMPO, a mitochondrial ROS scavenger. TRPM2 deficiency hinders cisplatin-induced autophagy via blockage of Ca2+ influx and subsequent up-regulation of AKT-mTOR signaling. Consistently, cisplatin-induced tubular mitochondrial damage, cell apoptosis and renal dysfunction in TRPM2-deficient mice are mitigated by treatment with a mTOR inhibitor. Conclusion: Our results suggest that the TRPM2 channel plays a protective role in cisplatin-induced AKI via modulating the Ca2+-AKT-mTOR signaling pathway and autophagy, providing novel insights into the pathogenesis of kidney injury.
Collapse
Affiliation(s)
- Binfeng Yu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
- Department of Infectious Disease, Sir Run Run Shaw Hospital, Zhejiang University School of medicine, Hangzhou 310003, China
| | - Lini Jin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xi Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| | - Yi Zhang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Gensheng Zhang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- The Children's Hospital, Zhejiang University School of medicine, Hangzhou 310003, China
| | - Fangqin Wang
- The Children's Hospital, Zhejiang University School of medicine, Hangzhou 310003, China
| | - Xinwan Su
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Qiuyuan Fang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Liang Xiao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| | - Yi Yang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, and Department of Physiology and Pathophysiology, Xinxiang Medical University, P.R. China
- A4245-Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, France
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| | - Wei Yang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Weiqiang Lin
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| |
Collapse
|
27
|
Bhatia D, Choi ME. Autophagy and mitophagy: physiological implications in kidney inflammation and diseases. Am J Physiol Renal Physiol 2023; 325:F1-F21. [PMID: 37167272 PMCID: PMC10292977 DOI: 10.1152/ajprenal.00012.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Autophagy is a ubiquitous intracellular cytoprotective quality control program that maintains cellular homeostasis by recycling superfluous cytoplasmic components (lipid droplets, protein, or glycogen aggregates) and invading pathogens. Mitophagy is a selective form of autophagy that by recycling damaged mitochondrial material, which can extracellularly act as damage-associated molecular patterns, prevents their release. Autophagy and mitophagy are indispensable for the maintenance of kidney homeostasis and exert crucial functions during both physiological and disease conditions. Impaired autophagy and mitophagy can negatively impact the pathophysiological state and promote its progression. Autophagy helps in maintaining structural integrity of the kidney. Mitophagy-mediated mitochondrial quality control is explicitly critical for regulating cellular homeostasis in the kidney. Both autophagy and mitophagy attenuate inflammatory responses in the kidney. An accumulating body of evidence highlights that persistent kidney injury-induced oxidative stress can contribute to dysregulated autophagic and mitophagic responses and cell death. Autophagy and mitophagy also communicate with programmed cell death pathways (apoptosis and necroptosis) and play important roles in cell survival by preventing nutrient deprivation and regulating oxidative stress. Autophagy and mitophagy are activated in the kidney after acute injury. However, their aberrant hyperactivation can be deleterious and cause tissue damage. The findings on the functions of autophagy and mitophagy in various models of chronic kidney disease are heterogeneous and cell type- and context-specific dependent. In this review, we discuss the roles of autophagy and mitophagy in the kidney in regulating inflammatory responses and during various pathological manifestations.
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, New York, 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, New York, United States
| |
Collapse
|
28
|
Shao YF, Tang BB, Ding YH, Fang CY, Hong L, Shao CX, Yang ZX, Qiu YP, Wang JC, Yang B, Weng QJ, Wang JJ, He QJ. Kaempferide ameliorates cisplatin-induced nephrotoxicity via inhibiting oxidative stress and inducing autophagy. Acta Pharmacol Sin 2023; 44:1442-1454. [PMID: 36658427 PMCID: PMC10310756 DOI: 10.1038/s41401-023-01051-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Acute kidney injury (AKI) caused by anti-tumor drugs, such as cisplatin, is a severe complication with no effective treatment currently, leading to the reduction or discontinuation of chemotherapy. Natural products or herbal medicines are gradually considered as promising agents against cisplatin-induced AKI with the advantages of multi-targeting, multi-effects, and less resistance. In this study, we investigated the effects of kaempferide, a natural flavonoid extracted from the rhizome of Kaempferia galanga, in experimental AKI models in vitro and in vivo. We first conducted pharmacokinetic study in mice and found a relative stable state of kaempferide with a small amount of conversion into kaempferol. We showed that both kaempferide (10 μM) and kaempferol (10 μM) significantly inhibited cisplatin-caused injuries in immortalized proximal tubule epithelial cell line HK-2. In AKI mice induced by injection of a single dose of cisplatin (15 mg/kg), oral administration of kaempferide (50 mg/kg) either before or after cisplatin injection markedly improved renal function, and ameliorated renal tissue damage. We demonstrated that kaempferide inhibited oxidative stress and induced autophagy in cisplatin-treated mice and HK-2 cells, thus increasing tubular cell viability and decreasing immune responses to attenuate the disease progression. In addition, treatment with kaempferide significantly ameliorated ischemia-reperfusion-induced renal injury in vitro and in vivo. We conclude that kaempferide is a promising natural product for treating various AKI. This study has great implications for promotion of its use in healthcare products, and help to break through the limited use of cisplatin in the clinic.
Collapse
Affiliation(s)
- Yan-Fei Shao
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Bing-Bing Tang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yu-Hui Ding
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chun-Yan Fang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ling Hong
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Chun-Xiao Shao
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhao-Xu Yang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue-Ping Qiu
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jin-Cheng Wang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qin-Jie Weng
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia-Jia Wang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Qiao-Jun He
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
29
|
Pan Z, Zhang H, Dokudovskaya S. The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics. Int J Mol Sci 2023; 24:10651. [PMID: 37445831 DOI: 10.3390/ijms241310651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Cisplatin (cis-diamminedichloroplatinum I) is a platinum-based drug, the mainstay of anticancer treatment for numerous solid tumors. Since its approval by the FDA in 1978, the drug has continued to be used for the treatment of half of epithelial cancers. However, resistance to cisplatin represents a major obstacle during anticancer therapy. Here, we review recent findings on how the mTORC1 pathway and autophagy can influence cisplatin sensitivity and resistance and how these data can be applicable for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zhenrui Pan
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Hanxiao Zhang
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Svetlana Dokudovskaya
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| |
Collapse
|
30
|
Zhang Y, Zeng M, Li B, Zhang B, Cao B, Wu Y, Ye S, Xu R, Zheng X, Feng W. Ephedra Herb extract ameliorates adriamycin-induced nephrotic syndrome in rats via the CAMKK2/AMPK/mTOR signaling pathway. Chin J Nat Med 2023; 21:371-382. [PMID: 37245875 DOI: 10.1016/s1875-5364(23)60454-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Indexed: 05/30/2023]
Abstract
This study aimed to investigate the effect and mechanisms of Ephedra Herb (EH) extract on adriamycin-induced nephrotic syndrome (NS), providing an experimental basis for the clinical treatment of NS. Hematoxylin and eosin staining, creatinine, urea nitrogen, and kidn injury molecule-1 were used to evaluate the activities of EH extract on renal function. The levels of inflammatory factors and oxidative stress were detected by kits. The levels of reactive oxygen species, immune cells, and apoptosis were measured by flow cytometry. A network pharmacological approach was used to predict the potential targets and mechanisms of EH extract in the treatment of NS. The protein levels of apoptosis-related proteins and CAMKK2, p-CAMKK2, AMPK, p-AMPK, mTOR and p-mTOR in the kidneys were detected by Western blot. The effective material basis of EH extract was screened by MTT assay. The AMPK pathway inhibitor (compound C, CC) was added to investigate the effect of the potent material basis on adriamycin-induced cell injury. EH extract significantly improved renal injury and relieve inflammation, oxidative stress, and apoptosis in rats. Network pharmacology and Western blot results showed that the effect of EH extract on NS may be associated with the CAMKK2/AMPK/mTOR signaling pathway. Moreover, methylephedrine significantly ameliorated adriamycin-induced NRK-52e cell injury. Methylephedrine also significantly improved the phosphorylation of AMPK and mTOR, which were blocked by CC. In sum, EH extract may ameliorate renal injury via the CAMKK2/AMPK/mTOR signaling pathway. Moreover, methylephedrine may be one of the material bases of EH extract.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., Zhengzhou 450000, China
| | - Benke Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Bing Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Yuanyuan Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Shan Ye
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Ruiqi Xu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., Zhengzhou 450000, China.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., Zhengzhou 450000, China.
| |
Collapse
|
31
|
Du YW, Li XK, Wang TT, Zhou L, Li HR, Feng L, Ma H, Liu HB. Cyanidin-3-glucoside inhibits ferroptosis in renal tubular cells after ischemia/reperfusion injury via the AMPK pathway. Mol Med 2023; 29:42. [PMID: 37013504 PMCID: PMC10069074 DOI: 10.1186/s10020-023-00642-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Ferroptosis, which is characterized by lipid peroxidation and iron accumulation, is closely associated with the pathogenesis of acute renal injury (AKI). Cyanidin-3-glucoside (C3G), a typical flavonoid that has anti-inflammatory and antioxidant effects on ischemia‒reperfusion (I/R) injury, can induce AMP-activated protein kinase (AMPK) activation. This study aimed to show that C3G exerts nephroprotective effects against I/R-AKI related ferroptosis by regulating the AMPK pathway. METHODS Hypoxia/reoxygenation (H/R)-induced HK-2 cells and I/R-AKI mice were treated with C3G with or without inhibiting AMPK. The level of intracellular free iron, the expression of the ferroptosis-related proteins acyl-CoA synthetase long chain family member 4 (ACSL4) and glutathione peroxidase 4 (GPX4), and the levels of the lipid peroxidation markers 4-hydroxynonenal (4-HNE), lipid reactive oxygen species (ROS) and malondialdehyde (MDA) were examined. RESULTS We observed the inhibitory effect of C3G on ferroptosis in vitro and in vivo, which was characterized by the reversion of excessive intracellular free iron accumulation, a decrease in 4-HNE, lipid ROS, MDA levels and ACSL4 expression, and an increase in GPX4 expression and glutathione (GSH) levels. Notably, the inhibition of AMPK by CC significantly abrogated the nephroprotective effect of C3G on I/R-AKI models in vivo and in vitro. CONCLUSION Our results provide new insight into the nephroprotective effect of C3G on acute I/R-AKI by inhibiting ferroptosis by activating the AMPK pathway.
Collapse
Affiliation(s)
- Yi-Wei Du
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Xiao-Kang Li
- Department of Cardiology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Ting-Ting Wang
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Lu Zhou
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Hui-Rong Li
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Lan Feng
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China.
| | - Hong-Bao Liu
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China.
| |
Collapse
|
32
|
Danics L, Abbas AA, Kis B, Pircs K. Fountain of youth—Targeting autophagy in aging. Front Aging Neurosci 2023; 15:1125739. [PMID: 37065462 PMCID: PMC10090449 DOI: 10.3389/fnagi.2023.1125739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
As our society ages inexorably, geroscience and research focusing on healthy aging is becoming increasingly urgent. Macroautophagy (referred to as autophagy), a highly conserved process of cellular clearance and rejuvenation has attracted much attention due to its universal role in organismal life and death. Growing evidence points to autophagy process as being one of the key players in the determination of lifespan and health. Autophagy inducing interventions show significant improvement in organismal lifespan demonstrated in several experimental models. In line with this, preclinical models of age-related neurodegenerative diseases demonstrate pathology modulating effect of autophagy induction, implicating its potential to treat such disorders. In humans this specific process seems to be more complex. Recent clinical trials of drugs targeting autophagy point out some beneficial effects for clinical use, although with limited effectiveness, while others fail to show any significant improvement. We propose that using more human-relevant preclinical models for testing drug efficacy would significantly improve clinical trial outcomes. Lastly, the review discusses the available cellular reprogramming techniques used to model neuronal autophagy and neurodegeneration while exploring the existing evidence of autophagy’s role in aging and pathogenesis in human-derived in vitro models such as embryonic stem cells (ESCs), induced pluripotent stem cell derived neurons (iPSC-neurons) or induced neurons (iNs).
Collapse
Affiliation(s)
- Lea Danics
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU), Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SU), Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
| | - Anna Anoir Abbas
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU), Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary
| | - Balázs Kis
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU), Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary
| | - Karolina Pircs
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU), Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
- *Correspondence: Karolina Pircs,
| |
Collapse
|
33
|
Zhou Q, Quirk JD, Hu Y, Yan H, Gaut JP, Pham CTN, Wickline SA, Pan H. Rapamycin Perfluorocarbon Nanoparticle Mitigates Cisplatin-Induced Acute Kidney Injury. Int J Mol Sci 2023; 24:6086. [PMID: 37047059 PMCID: PMC10093942 DOI: 10.3390/ijms24076086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
For nearly five decades, cisplatin has played an important role as a standard chemotherapeutic agent and been prescribed to 10-20% of all cancer patients. Although nephrotoxicity associated with platinum-based agents is well recognized, treatment of cisplatin-induced acute kidney injury is mainly supportive and no specific mechanism-based prophylactic approach is available to date. Here, we postulated that systemically delivered rapamycin perfluorocarbon nanoparticles (PFC NP) could reach the injured kidneys at sufficient and sustained concentrations to mitigate cisplatin-induced acute kidney injury and preserve renal function. Using fluorescence microscopic imaging and fluorine magnetic resonance imaging/spectroscopy, we illustrated that rapamycin-loaded PFC NP permeated and were retained in injured kidneys. Histologic evaluation and blood urea nitrogen (BUN) confirmed that renal structure and function were preserved 48 h after cisplatin injury. Similarly, weight loss was slowed down. Using western blotting and immunofluorescence staining, mechanistic studies revealed that rapamycin PFC NP significantly enhanced autophagy in the kidney, reduced the expression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), as well as decreased the expression of the apoptotic protein Bax, all of which contributed to the suppression of apoptosis that was confirmed with TUNEL staining. In summary, the delivery of an approved agent such as rapamycin in a PFC NP format enhances local delivery and offers a novel mechanism-based prophylactic therapy for cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Qingyu Zhou
- Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ying Hu
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Huimin Yan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph P. Gaut
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
34
|
Shen SH, Wang RL, Yuan Q, Jian LY, Guo HH, Li HS, Liu XP, Huang RF. The roles of AMPK/mTOR autophagy pathway in the acute kidney injury-induced acute lung injury. CHINESE J PHYSIOL 2023; 66:73-84. [PMID: 37082995 DOI: 10.4103/cjop.cjop-d-22-00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Acute kidney injury (AKI) is one of the most challenging clinical problems in kidney disease due to serious complications and high mortality rate, which can lead to acute lung injury (ALI) through inflammatory reactions and oxidative stress. Adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway has been reported to be involved in the development of renal ischemia-reperfusion through autophagy and it remains unclear whether AMPK/mTOR pathway has an effect on the AKI-induced ALI. In this study, we aimed to investigate the effects of autophagy-related AMPK/mTOR signaling pathway on inflammatory factors and oxidative stress in an AKI-induced ALI model. The 48 male Sprague-Dawley rats were divided into four groups randomly: (i) sham, (ii) ischemia/reperfusion injury (IRI), (iii) IRI + rapamycin (RA), and (iv) IRI + 3-methyladenine (3-MA). Unilateral flank incisions were made and right kidneys were excised. The left kidney was subjected to 60 min of ischemia followed by 12, 24, 48, and 72 h of reperfusion. The levels of Scr, blood urea nitrogen (BUN), Wet/Dry ratio, indexes of inflammation, and oxidative stress were assayed. Histological examinations were performed. The protein expression of AMPK, mTOR, LC3-II/LC3-I ratio, and Beclin-1, ULK1 was evaluated by western blotting and immunohistochemistry. Compared to the rats from the sham group, IRI rats showed significantly pulmonary damage after AKI with increased Scr, BUN, Wet/Dry ratio, indexes of inflammation, and oxidative stress. The expression of AMPK, LC3-II/LC3-I ratio, Beclin-1, and ULK1 and were increased, while p62 and mTOR were decreased. In addition, RA treatment significantly attenuated lung injury by promoting autophagy through the activation of the AMPK/mTOR pathway, and 3-MA treatment exhibited adverse effects inversely. Therefore, the activation of the AMPK/mTOR pathway after renal IRI induction could significantly attenuate kidney injury and following AKI-induced ALI by inducing autophagy, which alienates inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Si-Heng Shen
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ruo-Lin Wang
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qi Yuan
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lu-Yong Jian
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hua-Hui Guo
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - He-Sheng Li
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xue-Pin Liu
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ren-Fa Huang
- Department of Nephropathy, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
35
|
Niu L, Wang L, He X, Fan Q, Chen M, Qiao Y, Huang H, Lai S, Wan Q, Zhang Z, He M, He H. Renoprotective effects of ferulic acid mediated by AMPKα1 against lipopolysaccharide-induced damage. Int Immunopharmacol 2023; 115:109703. [PMID: 37724953 DOI: 10.1016/j.intimp.2023.109703] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
The kidney is susceptible to lipopolysaccharide (LPS)-induced damage with sepsis, and renal dysfunction is a leading cause of mortality in patients with sepsis. However, the renoprotective effects of ferulic acid (FA) during sepsis and the underlying mechanism remain unclear. This study explored these renoprotective effects using NRK-52E cells and mice with LPS-induced renal damage. The results showed that after LPS challenge, NRK-52E cell viability decreased, whereas lactate dehydrogenase, caspase-3 activity, apoptosis, the release of the inflammatory cytokines, and reactive oxygen species generation increased. Further, the activities of endogenous enzymatic and non-enzymatic antioxidant systems, and energy metabolism were inhibited, mitochondrial membrane potential was lost, mitochondrial permeability transition pores opened, renal blood flow and excretory functions were reduced, and the morphology and ultrastructure of renal tissue were seriously damaged in mice exposed to LPS. FA pretreatment upregulated AMP-activated protein kinase (AMPK) α1 expression and phosphorylation and significantly reversed the aforementioned functional, enzymological, and morphological indexes in vivo and in vitro. However, these renoprotective effects of FA were attenuated by compound C, an AMPK inhibitor. In conclusion, FA pretreatment can upregulate AMPKα1 expression and phosphorylation, inhibit inflammatory cytokine release and oxidative stress, improve mitochondrial function and energy supply, alleviate apoptosis, and ultimately protect renal tissue against LPS damage.
Collapse
Affiliation(s)
- Li Niu
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Liang Wang
- Department of Rehabilitation, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xinlan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qigui Fan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Maosi Chen
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Yang Qiao
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Huang Huang
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Songqing Lai
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Wan
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zeyu Zhang
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ming He
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| |
Collapse
|
36
|
Xiang Y, Fu Y, Wu W, Tang C, Dong Z. Autophagy in acute kidney injury and maladaptive kidney repair. BURNS & TRAUMA 2023; 11:tkac059. [PMID: 36694860 PMCID: PMC9867874 DOI: 10.1093/burnst/tkac059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/30/2022] [Accepted: 12/20/2022] [Indexed: 01/23/2023]
Abstract
Acute kidney injury (AKI) is a major renal disease characterized by a sudden decrease in kidney function. After AKI, the kidney has the ability to repair, but if the initial injury is severe the repair may be incomplete or maladaptive and result in chronic kidney problems. Autophagy is a highly conserved pathway to deliver intracellular contents to lysosomes for degradation. Autophagy plays an important role in maintaining renal function and is involved in the pathogenesis of renal diseases. Autophagy is activated in various forms of AKI and acts as a defense mechanism against kidney cell injury and death. After AKI, autophagy is maintained at a relatively high level in kidney tubule cells during maladaptive kidney repair but the role of autophagy in maladaptive kidney repair has been controversial. Nonetheless, recent studies have demonstrated that autophagy may contribute to maladaptive kidney repair after AKI by inducing tubular degeneration and promoting a profibrotic phenotype in renal tubule cells. In this review, we analyze the role and regulation of autophagy in kidney injury and repair and discuss the therapeutic strategies by targeting autophagy.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Wenwen Wu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | | |
Collapse
|
37
|
Chen J, Zhu G, Xiao W, Huang X, Wang K, Zong Y. Ginsenoside Rg1 Ameliorates Pancreatic Injuries via the AMPK/mTOR Pathway in vivo and in vitro. Diabetes Metab Syndr Obes 2023; 16:779-794. [PMID: 36945297 PMCID: PMC10024876 DOI: 10.2147/dmso.s401642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND The main propanaxatriol-type saponin found in ginseng (Panax ginseng C. A. Mey), ginsenoside Rg1 (G-Rg1), has bioactivities that include anti-inflammatory, antioxidant, and anti-diabetic properties. This study aimed to investigate the effects of G-Rg1 on streptozotocin (STZ)-induced Type 1 Diabetes mellitus (T1DM) mice and the insulin-secreting cell line in RIN-m5F cells with high-glucose (HG) treatment. METHODS The STZ-induced DM mice model was treated with G-Rg1 alone or combined with 3-Methyladenine (3-MA, an autophagy inhibitor)/rapamycin (RAPA, an autophagy activator) for 8 weeks, and levels of glucose and lipid metabolism, histopathological changes, as well as autophagy and apoptosis of relevant markers were estimated. In vitro, the HG-induced RIN-m5F cells were treated with G-Rg1, 3-MA, and Compound C (CC), an AMPK inhibitor, or their combinations to estimate the influences on cell apoptosis, autophagy, and AMPK/mTOR pathway-associated target gene levels. RESULTS G-Rg1 treatment attenuated glucose and lipid metabolism disorder and pancreatic fibrosis in diabetic mice. In addition, subdued autophagy and p-AMPK protein expression, and enhanced p-mTOR protein expression and apoptosis levels in TIDM mice and HG-induced RIN-m5F cells were ameliorated by G-Rg1 treatment. Furthermore, these anti-apoptosis effects of G-Rg1 were partially abolished by 3-MA and CC. CONCLUSION Our findings revealed that G-Rg1 exhibits strong anti-apoptosis ability in pancreatic tissues of type 1 diabetic mice and HG-induced RIN-m5F cells, and the mechanisms involved in activating AMPK and inhibiting mTOR-mediated autophagy, indicating that G-Rg1 may have the therapeutic and preventive potential for treating pancreatic injury in diabetic patients.
Collapse
Affiliation(s)
- Jin Chen
- Department of Hematology, Yiwu Central Hospital, Yiwu, People’s Republic of China
| | - Guoping Zhu
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
| | - Wenbo Xiao
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xiaosong Huang
- Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
| | - Kewu Wang
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
- Correspondence: Kewu Wang; Yi Zong, Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, No. N1, Shangcheng Avenue, Yiwu, Zhejiang, People’s Republic of China, Email ;
| | - Yi Zong
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
| |
Collapse
|
38
|
Wang H, Wang Q, Wu Y, Lou J, Zhu S, Xu Y. Autophagy-related gene LAPTM4B promotes the progression of renal clear cell carcinoma and is associated with immunity. Front Pharmacol 2023; 14:1118217. [PMID: 36937841 PMCID: PMC10017457 DOI: 10.3389/fphar.2023.1118217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is a common urologic disease. Currently, surgery is the primary treatment for renal cancer; immunotherapy is not as effective a treatment strategy as expected. Hence, understanding the mechanism in the tumor immune microenvironment (TME) and exploring novel immunotherapeutic targets are considered important. Recent studies have demonstrated that autophagy could affect the immune environment of renal cell carcinoma and induce proliferation and apoptosis of cancer cells. By comparing lysosomal genes and regulating autophagy genes, we identified the LAPTM4B gene to be related to RCC autophagy. By analyzing the TCGA-KIRC cohort using bioinformatics, we found M2 macrophages associated with tumor metastasis to be significantly increased in the immune microenvironment of patients with high expression of LAPTM4B. GO/KEGG/GSEA/GSVA results showed significant differences in tumor autophagy- and metastasis-related pathways. Single-cell sequencing was used to compare the expression of LAPTM4B in different cell types and obtain the differences in lysosomal and autophagy pathway activities in different ccRCC cells. Subsequently, we confirmed the differential expression of LAPTM4B in renal cell carcinoma of different Fuhrman grades using western blotting. Downregulation of LAPTM4B expression significantly reduced the proliferation of renal cell carcinoma cells and promoted cell apoptosis through cell experiments. Overall, our study demonstrated that the autophagy-related gene LAPTM4B plays a critical role in the TME of RCC, and suggested that LAPTM4B is a potential therapeutic target for RCC immunotherapy.
Collapse
Affiliation(s)
- He Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qibo Wang
- Department of Urology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Yaoyao Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianmin Lou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shaoxing Zhu
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
- *Correspondence: Shaoxing Zhu, ; Yipeng Xu,
| | - Yipeng Xu
- Department of Urology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- *Correspondence: Shaoxing Zhu, ; Yipeng Xu,
| |
Collapse
|
39
|
A Hydrodistillate of Gynostemma pentaphyllum and Damulin B Prevent Cisplatin-Induced Nephrotoxicity In Vitro and In Vivo via Regulation of AMPKα1 Transcription. Nutrients 2022; 14:nu14234997. [PMID: 36501027 PMCID: PMC9737728 DOI: 10.3390/nu14234997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
The clinical application of cisplatin, one of the most effective chemotherapeutic agents used to treat various cancers, has been limited by the risk of adverse effects, notably nephrotoxicity. Despite intensive research for decades, there are no effective approaches for alleviating cisplatin nephrotoxicity. This study aimed to investigate the protective effects and potential mechanisms of a Gynostemma pentaphyllum leaves hydrodistillate (GPHD) and its major component, damulin B, against cisplatin-induced nephrotoxicity in vitro and in vivo. A hydro-distillation method can extract large amounts of components within a short period of time using non-toxic, environmentally friendly solvent. We found that the levels of AMP-activated protein kinase α1 (AMPKα1), reactive oxygen species (ROS), and apoptosis were tightly associated with each other in HEK293 cells treated with cisplatin. We demonstrated that AMPKα1 acted as an anti-oxidant factor and that ROS generated by cisplatin suppressed the expression of AMPKα1 at the transcriptional level, thereby resulting in induction of apoptosis. Treatment with GPHD or damulin B effectively prevented cisplatin-induced apoptosis of HEK293 cells and cisplatin-induced acute kidney injury in mice by suppressing oxidative stress and maintaining AMPKα1 levels. Therefore, our study suggests that GPHD and damulin B may serve as prospective adjuvant agents against cisplatin-induced nephrotoxicity.
Collapse
|
40
|
Al-kuraishy HM, Al-Gareeb AI, Elekhnawy E, Batiha GES. Nitazoxanide and COVID-19: A review. Mol Biol Rep 2022; 49:11169-11176. [PMID: 36094778 PMCID: PMC9465141 DOI: 10.1007/s11033-022-07822-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/26/2022] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a current global illness triggered by severe acute respiratory coronavirus 2 (SARS-CoV-2) leading to acute viral pneumonia, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and cytokine storm in severe cases. In the COVID-19 era, different unexpected old drugs are repurposed to find out effective and cheap therapies against SARS-CoV-2. One of these elected drugs is nitazoxanide (NTZ) which is an anti-parasitic drug with potent antiviral activity. It is effectively used in the treatment of protozoa and various types of helminths in addition to various viral infections. Thus, we aimed to elucidate the probable effect of NTZ on SARS-CoV-2 infections. Findings of the present study illustrated that NTZ can reduce SARS-CoV-2-induced inflammatory reactions through activation of interferon (IFN), restoration of innate immunity, inhibition of the release of pro-inflammatory cytokines, suppression of the mammalian target of rapamycin (mTOR), and induction of autophagic cell death. Moreover, it can inhibit the induction of oxidative stress which causes cytokine storm and is associated with ALI, ARDS, and multi-organ damage (MOD). This study concluded that NTZ has important anti-inflammatory and immunological properties that may mitigate SARS-CoV-2 infection-induced inflammatory disorders. Despite broad-spectrum antiviral properties of NTZ, the direct anti-SARS-CoV-2 effect was not evident and documented in recent studies. Then, in silico and in vitro studies in addition to clinical trials and prospective studies are needed to confirm the beneficial impact of NTZ on the pathogenesis of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Ali I. Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| |
Collapse
|
41
|
The oncoprotein MUC1 facilitates breast cancer progression by promoting Pink1-dependent mitophagy via ATAD3A destabilization. Cell Death Dis 2022; 13:899. [PMID: 36289190 PMCID: PMC9606306 DOI: 10.1038/s41419-022-05345-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
Mitophagy is a vital process that controls mitochondria quality, dysregulation of which can promote cancer. Oncoprotein mucin 1 (MUC1) targets mitochondria to attenuate drug-induced apoptosis. However, little is known about whether and how MUC1 contributes to mitochondrial homeostasis in cancer cells. We identified a novel role of MUC1 in promoting mitophagy. Increased mitophagy is coupled with the translocation of MUC1 to mitochondria, where MUC1 interacts with and induces degradation of ATPase family AAA domain-containing 3A (ATAD3A), resulting in protection of PTEN-induced kinase 1 (Pink1) from ATAD3A-mediated cleavage. Interestingly, MUC1-induced mitophagy is associated with increased oncogenicity of cancer cells. Similarly, inhibition of mitophagy significantly suppresses MUC1-induced cancer cell activity in vitro and in vivo. Consistently, MUC1 and ATAD3A protein levels present an inverse relationship in tumor tissues of breast cancer patients. Our data validate that MUC1/ATAD3A/Pink1 axis-mediated mitophagy constitutes a novel mechanism for maintaining the malignancy of cancer cells, providing a novel therapeutic approach for MUC1-positive cancers.
Collapse
|
42
|
Synthetic Non-Coding RNA for Suppressing mTOR Translation to Prevent Renal Fibrosis Related to Autophagy in UUO Mouse Model. Int J Mol Sci 2022; 23:ijms231911365. [PMID: 36232665 PMCID: PMC9569483 DOI: 10.3390/ijms231911365] [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: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022] Open
Abstract
The global burden of chronic kidney disease is increasing, and the majority of these diseases are progressive. Special site-targeted drugs are emerging as alternatives to traditional drugs. Oligonucleotides (ODNs) have been proposed as effective therapeutic tools in specific molecular target therapies for several diseases. We designed ring-type non-coding RNAs (ncRNAs), also called mTOR ODNs to suppress mammalian target rapamycin (mTOR) translation. mTOR signaling is associated with excessive cell proliferation and fibrogenesis. In this study, we examined the effects of mTOR suppression on chronic renal injury. To explore the regulation of fibrosis and inflammation in unilateral ureteral obstruction (UUO)-induced injury, we injected synthesized ODNs via the tail vein of mice. The expression of inflammatory-related markers (interleukin-1β, tumor necrosis factor-α), and that of fibrosis (α-smooth muscle actin, fibronectin), was decreased by synthetic ODNs. Additionally, ODN administration inhibited the expression of autophagy-related markers, microtubule-associated protein light chain 3, Beclin1, and autophagy-related gene 5-12. We confirmed that ring-type ODNs inhibited fibrosis, inflammation, and autophagy in a UUO mouse model. These results suggest that mTOR may be involved in the regulation of autophagy and fibrosis and that regulating mTOR signaling may be a therapeutic strategy against chronic renal injury.
Collapse
|
43
|
Fan Y, Li X, Sun H, Gao Z, Zhu Z, Yuan K. Role of WTAP in Cancer: From Mechanisms to the Therapeutic Potential. Biomolecules 2022; 12:biom12091224. [PMID: 36139062 PMCID: PMC9496264 DOI: 10.3390/biom12091224] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Wilms' tumor 1-associating protein (WTAP) is required for N6-methyladenosine (m6A) RNA methylation modifications, which regulate biological processes such as RNA splicing, cell proliferation, cell cycle, and embryonic development. m6A is the predominant form of mRNA modification in eukaryotes. WTAP exerts m6A modification by binding to methyltransferase-like 3 (METTL3) in the nucleus to form the METTL3-methyltransferase-like 14 (METTL14)-WTAP (MMW) complex, a core component of the methyltransferase complex (MTC), and localizing to the nuclear patches. Studies have demonstrated that WTAP plays a critical role in various cancers, both dependent and independent of its role in m6A modification of methyltransferases. Here, we describe the recent findings on the structural features of WTAP, the mechanisms by which WTAP regulates the biological functions, and the molecular mechanisms of its functions in various cancers. By summarizing the latest WTAP research, we expect to provide new directions and insights for oncology research and discover new targets for cancer treatment.
Collapse
Affiliation(s)
- Yongfei Fan
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Xinwei Li
- Department of Gastroenterology, Affiliated Cancer Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Huihui Sun
- Department of Radiotherapy, The Affiliated Changzhou No. 1 People’s Hospital of Suzhou University, Changzhou 213003, China
| | - Zhaojia Gao
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Zheng Zhu
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Kai Yuan
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213003, China
- Correspondence:
| |
Collapse
|
44
|
Liu W, Gan Y, Ding Y, Zhang L, Jiao X, Liu L, Cao H, Gu Y, Yan L, Wang Y, Wang L, Chen S, Shao F. Autophagy promotes GSDME-mediated pyroptosis via intrinsic and extrinsic apoptotic pathways in cobalt chloride-induced hypoxia reoxygenation-acute kidney injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113881. [PMID: 35863214 DOI: 10.1016/j.ecoenv.2022.113881] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
Cobalt is a transition element that abundantly exists in the environment. Besides direct hypoxia stress, cobalt ions indirectly induce hypoxia-reoxygenation injury (HRI), the main cause of acute kidney injury (AKI), a life-threatening clinical syndrome characterized by the necrosis of the proximal tubular epithelial cells (PTECs) and inflammation. Pyroptosis, a type of inflammatory programmed cell death, might play an essential role in HRI-AKI. However, whether pyroptosis is involved in cobalt chloride (CoCl2)-induced HRI-AKI remains unknown. Autophagy is a cellular biological process maintaining cell homeostasis that is involved in cell damage in AKI, yet the underlying regulatory mechanism of autophagy on pyroptosis has not been fully understood. In this study, the in vitro and in vivo models of CoCl2-induced HRI-AKI were established with HK-2 cell line and C57BL/6J mouse. Pyroptosis-related markers were detected with western blotting and immunofluorescence assays, and results showed that gasdermin E (GSDME)-mediated pyroptosis was involved in the cell damage in HRI-AKI. Specific chemical inhibitors of caspase 3, caspase 8, and caspase 9 significantly inhibited GSDME-mediated pyroptosis, verifying that GSDME-mediated pyroptosis was induced via the activation of caspase 3/8/9. The western blotting and immunofluorescence assays were adopted to detect the accumulation of the autophagosomes, and results suggested that HRI increased the autophagic level. The effects of autophagy on apoptosis and pyroptosis were evaluated using lentivirus transfection assays to knock down autophagy-specific genes atg5 and fip200, and results demonstrated that autophagy induced GSDME-mediated pyroptosis via apoptotic pathways in HRI-AKI. Our results revealed the involvement of GSDME-mediated pyroptosis in CoCl2-induced HRI-AKI and promoted the understanding of the regulatory mechanism of GSDME cleavage. Our study might provide a potential therapeutic target for HRI-AKI, and will be helpful for the risk evaluation of cobalt exposure.
Collapse
Affiliation(s)
- Wenna Liu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yujin Gan
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yun Ding
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lina Zhang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Xiaojing Jiao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Lu Liu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Huixia Cao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Yue Gu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Lei Yan
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Yanliang Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China
| | - Limeng Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China.
| | - Song Chen
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450053, China.
| | - Fengmin Shao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, 7 Weiwu Road, Henan 450053, China.
| |
Collapse
|
45
|
Harley G, Katerelos M, Gleich K, de Souza DP, Narayana VK, Kemp BE, Power DA, Mount PF. Blocking AMPK signalling to acetyl-CoA carboxylase increases cisplatin-induced acute kidney injury and suppresses the benefit of metformin. Biomed Pharmacother 2022; 153:113377. [DOI: 10.1016/j.biopha.2022.113377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/25/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022] Open
|
46
|
Yang J, Yuan L, Liu F, Li L, Liu J, Chen Y, Lu Y, Yuan Y. Molecular mechanisms and physiological functions of autophagy in kidney diseases. Front Pharmacol 2022; 13:974829. [PMID: 36081940 PMCID: PMC9446454 DOI: 10.3389/fphar.2022.974829] [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: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 12/04/2022] Open
Abstract
Autophagy is a highly conserved cellular progress for the degradation of cytoplasmic contents including micromolecules, misfolded proteins, and damaged organelles that has recently captured attention in kidney diseases. Basal autophagy plays a pivotal role in maintaining cell survival and kidney homeostasis. Accordingly, dysregulation of autophagy has implicated in the pathologies of kidney diseases. In this review, we summarize the multifaceted role of autophagy in kidney aging, maladaptive repair, tubulointerstitial fibrosis and discuss autophagy-related drugs in kidney diseases. However, uncertainty still remains as to the precise mechanisms of autophagy in kidney diseases. Further research is needed to clarify the accurate molecular mechanism of autophagy in kidney diseases, which will facilitate the discovery of a promising strategy for the prevention and treatment of kidney diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanrong Lu
- *Correspondence: Yanrong Lu, ; Yujia Yuan,
| | - Yujia Yuan
- *Correspondence: Yanrong Lu, ; Yujia Yuan,
| |
Collapse
|
47
|
Li X, Yao Y, Wang J, Shen Z, Jiang Z, Xu S. Eucalyptol relieves imidacloprid-induced autophagy through the miR-451/Cab39/AMPK axis in Ctenopharyngodon idellus kidney cells †. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 249:106204. [PMID: 35661494 DOI: 10.1016/j.aquatox.2022.106204] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 05/14/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Imidacloprid (IMI) is a widely used neonicotinoid insecticide that has toxic effects on nontarget organisms. 1,8-Cineole (eucalyptol) is purified from essential oils in several aromatic plants and can prevent xenobiotic toxicity. The kidney is a major organ for xenobiotic elimination and thus has high risk of exposure. The purpose of this research was to clarify the effect of IMI exposure on autophagy in fish kidney cells, determine the potential of eucalyptol to provide cytoprotection from the toxicity of the neonicotinoid pesticide IMI, and identify its mechanism of action. Therefore, the Ctenopharyngodon idellus kidney cell line (CIK cell) was treated with 20 mg/L IMI and/or 20 μM eucalyptol for 48 h as the research objective. The results showed that IMI exposure induced autophagy accompanied by advanced autophagy markers BNIP3, Beclin1 and LC3Ⅱ/Ⅰ in CIK cells, reduced the levels of miR-451, increased the expression of Cab39 and AMPK, inhibited AKT/mTOR signaling, and activated the JNK pathway. Eucalyptol treatment alleviated IMI-induced autophagy and relieved the activation of autophagy-associated signals. These results indicate that eucalyptol could alleviate IMI-induced autophagy through the miR-451/Cab39/AMPK axis in fish kidney cells. These results partly explained the mechanism of biological threat on fish under IMI exposure and the potential application value of EUC in aquaculture.
Collapse
Affiliation(s)
- Xiaojing Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yujie Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jinliang Wang
- Shandong Binzhou Anim Sci & Vet Med Acad, Binzhou, 256600, Shandong, PR China
| | - Zhiqiang Shen
- Shandong Binzhou Anim Sci & Vet Med Acad, Binzhou, 256600, Shandong, PR China
| | - Zhihui Jiang
- Anyang Inst Technol, Henan Joint Int Res Lab Vet Biol Res & Applicat, Anyang, 455000, Henan, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
48
|
Vitto VAM, Bianchin S, Zolondick AA, Pellielo G, Rimessi A, Chianese D, Yang H, Carbone M, Pinton P, Giorgi C, Patergnani S. Molecular Mechanisms of Autophagy in Cancer Development, Progression, and Therapy. Biomedicines 2022; 10:biomedicines10071596. [PMID: 35884904 PMCID: PMC9313210 DOI: 10.3390/biomedicines10071596] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is an evolutionarily conserved and tightly regulated process that plays an important role in maintaining cellular homeostasis. It involves regulation of various genes that function to degrade unnecessary or dysfunctional cellular components, and to recycle metabolic substrates. Autophagy is modulated by many factors, such as nutritional status, energy level, hypoxic conditions, endoplasmic reticulum stress, hormonal stimulation and drugs, and these factors can regulate autophagy both upstream and downstream of the pathway. In cancer, autophagy acts as a double-edged sword depending on the tissue type and stage of tumorigenesis. On the one hand, autophagy promotes tumor progression in advanced stages by stimulating tumor growth. On the other hand, autophagy inhibits tumor development in the early stages by enhancing its tumor suppressor activity. Moreover, autophagy drives resistance to anticancer therapy, even though in some tumor types, its activation induces lethal effects on cancer cells. In this review, we summarize the biological mechanisms of autophagy and its dual role in cancer. In addition, we report the current understanding of autophagy in some cancer types with markedly high incidence and/or lethality, and the existing therapeutic strategies targeting autophagy for the treatment of cancer.
Collapse
Affiliation(s)
- Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Silvia Bianchin
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Alicia Ann Zolondick
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI 96816, USA; (A.A.Z.); (H.Y.); (M.C.)
- Department of Molecular Biosciences and Bioengineering, University of Hawai’i at Manoa, Honolulu, HI 96816, USA
| | - Giulia Pellielo
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Alessandro Rimessi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Diego Chianese
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI 96816, USA; (A.A.Z.); (H.Y.); (M.C.)
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI 96816, USA; (A.A.Z.); (H.Y.); (M.C.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
- Correspondence: (C.G.); (S.P.)
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
- Correspondence: (C.G.); (S.P.)
| |
Collapse
|
49
|
Li B, Lin F, Xia Y, Ye Z, Yan X, Song B, Yuan T, Li L, Zhou X, Yu W, Cheng F. The Intersection of Acute Kidney Injury and Non-Coding RNAs: Inflammation. Front Physiol 2022; 13:923239. [PMID: 35755446 PMCID: PMC9218900 DOI: 10.3389/fphys.2022.923239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022] Open
Abstract
Acute renal injury (AKI) is a complex clinical syndrome, involving a series of pathophysiological processes, in which inflammation plays a key role. Identification and verification of gene signatures associated with inflammatory onset and progression are imperative for understanding the molecular mechanisms involved in AKI pathogenesis. Non-coding RNAs (ncRNAs), involved in epigenetic modifications of inflammatory responses, are associated with the aberrant expression of inflammation-related genes in AKI. However, its regulatory role in gene expression involves precise transcriptional regulation mechanisms which have not been fully elucidated in the complex and volatile inflammatory response of AKI. In this study, we systematically review current research on the intrinsic molecular mechanisms of ncRNAs that regulate the inflammatory response in AKI. We aim to provide potential research directions and strategies for developing ncRNA-targeted gene therapies as an intervention for the inflammatory damage in AKI.
Collapse
Affiliation(s)
- Bojun Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinzhou Yan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baofeng Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianhui Yuan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
50
|
Numb Promotes Autophagy through p53 Pathway in Acute Kidney Injury Induced by Cisplatin. Anal Cell Pathol (Amst) 2022; 2022:8213683. [PMID: 35795076 PMCID: PMC9252835 DOI: 10.1155/2022/8213683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/14/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is an important public health concern and characterized as tubular death involved in apoptosis and necrosis. Autophagy is rapidly induced in tubules and associates with renal tubular cells homeostasis to have a complex link with tubular death in AKI. Numb is a multifunctional protein and exerts protective role in tubular death in AKI induced by Cisplatin. However, the effect of Numb on tubular autophagy remains to be investigated. In the present study, the protein expression of LC3 and Beclin-1 related to autophagy was analyzed in Cisplatin-induced AKI mice with knocking down Numb. In model of tubular injury induced by Cisplatin in vitro, downregulation of Numb in NRK-52E cells also inhibited the activation of autophagy accompanied with the decreased protein level of p53. Overexpression of Numb in NRK-52E cells activated autophagy with increased LC3 and Beclin-1 expression accompanied with increased protein level of p53. Moreover, autophagy activation following Numb overexpression was suppressed by p53 inhibitor pifithrin-α. These data indicate that Numb promotes p53-mediated activation of tubular autophagy in AKI induced by Cisplatin and therefore may provide important targets for the treatment of AKI.
Collapse
|