1
|
Shreya S, Dagar N, Shelke V, Puri B, Gaikwad AB. ULK1 as a therapeutic target in kidney diseases: Current perspective. Expert Opin Ther Targets 2024; 28:911-922. [PMID: 39526701 DOI: 10.1080/14728222.2024.2421762] [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: 01/13/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Globally, ~850 million people are affected by different kidney diseases. The pathogenesis of kidney diseases is intricate, where autophagy is crucial for maintaining kidney homeostasis. Iteliminates damaged organelles, thus reducing renal lesions and allowing tissue regeneration. Therefore, targeting various autophagy proteins, e.g. Unc-51-like autophagy-activating kinase 1 (ULK1), is emerging as potential therapeutic strategy against kidney disease. AREAS COVERED This review provides insights into the role of ULK1 as a therapeutic target in kidney diseases. Additionally, we have discussed the recent evidence based on pre-clinical studies for possible novel therapies modulating ULK1-mediated autophagy in kidney diseases. EXPERT OPINION ULK1 is one of the critical regulators of autophagy. Moreover, ULK1 works differently for different types of kidney disease. Considering its significant role in kidney disease pathogenesis, it could be a potential target to tackle kidney diseases. However, the dynamic molecular understanding of ULK1 in the context of various kidney diseases is still in its infancy and should be investigated further.
Collapse
Affiliation(s)
- Shruti Shreya
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Neha Dagar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Vishwadeep Shelke
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Bhupendra Puri
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | | |
Collapse
|
2
|
Noh MR, Padanilam BJ. Cell death induced by acute renal injury: a perspective on the contributions of accidental and programmed cell death. Am J Physiol Renal Physiol 2024; 327:F4-F20. [PMID: 38660714 PMCID: PMC11390133 DOI: 10.1152/ajprenal.00275.2023] [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: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
The involvement of cell death in acute kidney injury (AKI) is linked to multiple factors including energy depletion, electrolyte imbalance, reactive oxygen species, inflammation, mitochondrial dysfunction, and activation of several cell death pathway components. Since our review in 2003, discussing the relative contributions of apoptosis and necrosis, several other forms of cell death have been identified and are shown to contribute to AKI. Currently, these various forms of cell death can be fundamentally divided into accidental cell death and regulated or programmed cell death based on functional aspects. Several death initiator and effector molecules switch molecules that may act as signaling components triggering either death or protective mechanisms or alternate cell death pathways have been identified as part of the machinery. Intriguingly, several of these cell death pathways share components and signaling pathways suggesting complementary or compensatory functions. Thus, defining the cross talk between distinct cell death pathways and identifying the unique molecular effectors for each type of cell death may be required to develop novel strategies to prevent cell death. Furthermore, depending on the multiple forms of cell death simultaneously induced in different AKI settings, strategies for combination therapies that block multiple cell death pathways need to be developed to completely prevent injury, cell death, and renal function. This review highlights the various cell death pathways, cross talk, and interactions between different cell death modalities in AKI.
Collapse
Affiliation(s)
- Mi Ra Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Babu J Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|
3
|
Costigan C, Balgobin S, Zappitelli M. Drugs in treating paediatric acute kidney injury. Pediatr Nephrol 2023; 38:3923-3936. [PMID: 37052689 DOI: 10.1007/s00467-023-05956-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Acute kidney injury (AKI) is a complex syndrome which affects a significant proportion of hospitalized children. The breadth and impact of AKI on health outcomes in both adults and children have come to the fore in recent years with increasing awareness encouraging research advancement. Despite this, management strategies for most types of AKI remain heavily reliant on fluid and electrolyte management, hemodynamic optimization, nephrotoxin avoidance and appropriate initiation of kidney replacement therapy. Specific drugs targeting the mechanisms involved in AKI remain elusive. Recent improvement in appreciation of the complexity of AKI pathophysiology has allowed for greater opportunity to consider novel therapeutic agents. A number of drugs specifically targeting AKI are in various stages of development. This review will consider some novel and repurposed agents; interrogate the plausibility of the proposed mechanisms of action, as they relate to what we know about the pathophysiology of AKI; and review the level of existing literature supporting their efficacy. The evidence base, particularly in children, is limited.
Collapse
Affiliation(s)
- Caoimhe Costigan
- Department of Pediatrics, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Steve Balgobin
- Department of Pediatrics, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Michael Zappitelli
- Department of Pediatrics, Division of Nephrology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
- Peter Gilgan Centre for Research and Learning, 686 Bay Street, 11th floor, Rm 11.9722, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
4
|
Ai S, Li Y, Zheng H, Wang Z, Liu W, Tao J, Li Y, Wang Y. Global research trends and hot spots on autophagy and kidney diseases: a bibliometric analysis from 2000 to 2022. Front Pharmacol 2023; 14:1275792. [PMID: 38099142 PMCID: PMC10719858 DOI: 10.3389/fphar.2023.1275792] [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: 08/10/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Background: Autophagy is an essential cellular process involving the self-degradation and recycling of organelles, proteins, and cellular debris. Recent research has shown that autophagy plays a significant role in the occurrence and development of kidney diseases. However, there is a lack of bibliometric analysis regarding the relationship between autophagy and kidney diseases. Methods: A bibliometric analysis was conducted by searching for literature related to autophagy and kidney diseases in the Web of Science Core Collection (WoSCC) database from 2000 to 2022. Data processing was carried out using R package "Bibliometrix", VOSviewers, and CiteSpace. Results: A total of 4,579 articles related to autophagy and kidney diseases were collected from various countries. China and the United States were the main countries contributing to the publications. The number of publications in this field showed a year-on-year increasing trend, with open-access journals playing a major role in driving the literature output. Nanjing Medical University in China, Osaka University in Japan, and the University of Pittsburgh in the United States were the main research institutions. The journal "International journal of molecular sciences" had the highest number of publications, while "Autophagy" was the most influential journal in the field. These articles were authored by 18,583 individuals, with Dong, Zheng; Koya, Daisuke; and Kume, Shinji being the most prolific authors, and Dong, Zheng being the most frequently co-cited author. Research on autophagy mainly focused on diabetic kidney diseases, acute kidney injury, and chronic kidney disease. "Autophagy", "apoptosis", and "oxidative stress" were the primary research hotspots. Topics such as "diabetic kidney diseases", "sepsis", "ferroptosis", "nrf2", "hypertension" and "pi3k" may represent potential future development trends. Research on autophagy has gradually focused on metabolic-related kidney diseases such as diabetic nephropathy and hypertension. Additionally, PI3K, NRF2, and ferroptosis have been recent research directions in the field of autophagy mechanisms. Conclusion: This is the first comprehensive bibliometric study summarizing the relationship between autophagy and kidney diseases. The findings aid in identifying recent research frontiers and hot topics, providing valuable references for scholars investigating the role of autophagy in kidney diseases.
Collapse
Affiliation(s)
- Sinan Ai
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huijuan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - JiaYin Tao
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaotan Li
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaoxian Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
5
|
Ala M. Sestrin2 Signaling Pathway Regulates Podocyte Biology and Protects against Diabetic Nephropathy. J Diabetes Res 2023; 2023:8776878. [PMID: 36818747 PMCID: PMC9937769 DOI: 10.1155/2023/8776878] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023] Open
Abstract
Sestrin2 regulates cell homeostasis and is an upstream signaling molecule for several signaling pathways. Sestrin2 leads to AMP-activated protein kinase- (AMPK-) and GTPase-activating protein activity toward Rags (GATOR) 1-mediated inhibition of mammalian target of rapamycin complex 1 (mTORC1), thereby enhancing autophagy. Sestrin2 also improves mitochondrial biogenesis via AMPK/Sirt1/peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) signaling pathway. Blockade of ribosomal protein synthesis and augmentation of autophagy by Sestrin2 can prevent misfolded protein accumulation and attenuate endoplasmic reticulum (ER) stress. In addition, Sestrin2 enhances P62-mediated autophagic degradation of Keap1 to release nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 release by Sestrin2 vigorously potentiates antioxidant defense in diabetic nephropathy. Impaired autophagy and mitochondrial biogenesis, severe oxidative stress, and ER stress are all deeply involved in the development and progression of diabetic nephropathy. It has been shown that Sestrin2 expression is lower in the kidney of animals and patients with diabetic nephropathy. Sestrin2 knockdown aggravated diabetic nephropathy in animal models. In contrast, upregulation of Sestrin2 enhanced autophagy, mitophagy, and mitochondrial biogenesis and suppressed oxidative stress, ER stress, and apoptosis in diabetic nephropathy. Consistently, overexpression of Sestrin2 ameliorated podocyte injury, mesangial proliferation, proteinuria, and renal fibrosis in animal models of diabetic nephropathy. By suppressing transforming growth factor beta (TGF-β)/Smad and Yes-associated protein (YAP)/transcription enhancer factor 1 (TEF1) signaling pathways in experimental models, Sestrin2 hindered epithelial-mesenchymal transition and extracellular matrix accumulation in diabetic kidneys. Moreover, modulation of the downstream molecules of Sestrin2, for instance, augmentation of AMPK or Nrf2 signaling and inhibition of mTORC1, has been protective in diabetic nephropathy. Regarding the beneficial effects of Sestrin2 on diabetic nephropathy and its interaction with several signaling molecules, it is worth targeting Sestrin2 in diabetic nephropathy.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
6
|
Alsayed HA, Mohammad HMF, Khalil CM, El-Kherbetawy MK, Elaidy SM. Autophagy modulation by irbesartan mitigates the pulmonary fibrotic alterations in bleomycin challenged rats: Comparative study with rapamycin. Life Sci 2022; 303:120662. [PMID: 35636582 DOI: 10.1016/j.lfs.2022.120662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 01/07/2023]
Abstract
AIMS In pulmonary fibrosis, autophagy handles the maintenance of alveolar epithelial cells, prevents epithelial-mesenchymal transition (EMT), and controls collagen turnover. The mammalian target of rapamycin (mTOR) and its translational-dependent proteins are essential regulators of autophagy. Irbesartan (IRB) has earlier ameliorative effects in experimental pulmonary fibrosis. The current study aimed to explore therapeutic autophagy-modulated pulmonary fibrotic changes by IRB versus rapamycin (RAPA) in bleomycin (BLM)-challenged rats. MATERIALS AND METHODS A single intratracheal BLM dose at day (0), IRB in different doses (10, 20, and 40 mg/kg) or RAPA (2.5 mg/kg) was given daily for 14 continuous days. KEY FINDINGS IRB significantly diminished the fibrotic lung scores. Pulmonary levels of transforming growth factor (TGF)-β1 and hydroxyproline exhibited marked attenuation in IRB (40 mg/kg)-treated rats compared to other treated groups. IRB (40 mg/kg) was not significantly different from RAPA. It downregulated the fibrotic lung phosphorylated mammalian target of rapamycin (p-mTOR) levels and augmented lung Unc-51-like autophagy activating kinase 1 (ULK1), LC3-I and LC3-II more than IRB (10 and 20 mg/kg)-treated fibrotic groups. SIGNIFICANCE Autophagic effects via the mTOR signalling pathway may play a role in IRB's antifibrotic effects. Consideration of IRB as a therapeutic antifibrotic agent in pulmonary fibrosis needs further experimental and clinical long-term validation, especially in comorbid with primary hypertension, heart failure, and diabetic renal insults.
Collapse
Affiliation(s)
- Hadeer A Alsayed
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Hala M F Mohammad
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt; Central Laboratory, Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Cherine M Khalil
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | | | - Samah M Elaidy
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt.
| |
Collapse
|
7
|
Natural products: potential treatments for cisplatin-induced nephrotoxicity. Acta Pharmacol Sin 2021; 42:1951-1969. [PMID: 33750909 PMCID: PMC8633358 DOI: 10.1038/s41401-021-00620-9] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Cisplatin is a clinically advanced and highly effective anticancer drug used in the treatment of a wide variety of malignancies, such as head and neck, lung, testis, ovary, breast cancer, etc. However, it has only a limited use in clinical practice due to its severe adverse effects, particularly nephrotoxicity; 20%–35% of patients develop acute kidney injury (AKI) after cisplatin administration. The nephrotoxic effect of cisplatin is cumulative and dose dependent and often necessitates dose reduction or withdrawal. Recurrent episodes of AKI result in impaired renal tubular function and acute renal failure, chronic kidney disease, uremia, and hypertensive nephropathy. The pathophysiology of cisplatin-induced AKI involves proximal tubular injury, apoptosis, oxidative stress, inflammation, and vascular injury in the kidneys. At present, there are no effective drugs or methods for cisplatin-induced kidney injury. Recent in vitro and in vivo studies show that numerous natural products (flavonoids, saponins, alkaloids, polysaccharide, phenylpropanoids, etc.) have specific antioxidant, anti-inflammatory, and anti-apoptotic properties that regulate the pathways associated with cisplatin-induced kidney damage. In this review we describe the molecular mechanisms of cisplatin-induced nephrotoxicity and summarize recent findings in the field of natural products that undermine these mechanisms to protect against cisplatin-induced kidney damage and provide potential strategies for AKI treatment.
Collapse
|
8
|
Fukushima K, Kitamura S, Tsuji K, Wada J. Sodium-Glucose Cotransporter 2 Inhibitors Work as a "Regulator" of Autophagic Activity in Overnutrition Diseases. Front Pharmacol 2021; 12:761842. [PMID: 34744742 PMCID: PMC8566701 DOI: 10.3389/fphar.2021.761842] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Several large clinical trials have shown renal and cardioprotective effects of sodium–glucose cotransporter 2 (SGLT2) inhibitors in diabetes patients, and the protective mechanisms need to be elucidated. There have been accumulating studies which report that SGLT2 inhibitors ameliorate autophagy deficiency of multiple organs. In overnutrition diseases, SGLT2 inhibitors affect the autophagy via various signaling pathways, including mammalian target of rapamycin (mTOR), sirtuin 1 (SIRT1), and hypoxia-inducible factor (HIF) pathways. Recently, it turned out that not only stagnation but also overactivation of autophagy causes cellular damages, indicating that therapeutic interventions which simply enhance or stagnate autophagy activity might be a “double-edged sword” in some situations. A small number of studies suggest that SGLT2 inhibitors not only activate but also suppress the autophagy flux depending on the situation, indicating that SGLT2 inhibitors can “regulate” autophagic activity and help achieve the appropriate autophagy flux in each organ. Considering the complicated control and bilateral characteristics of autophagy, the potential of SGLT2 inhibitors as the regulator of autophagic activity would be beneficial in the treatment of autophagy deficiency.
Collapse
Affiliation(s)
- Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Academic Field of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Academic Field of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Academic Field of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Academic Field of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
9
|
Li T, Chen Y, Li Y, Yao Z, Liu W. FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice. Aging (Albany NY) 2021; 13:13535-13547. [PMID: 33819192 PMCID: PMC8202901 DOI: 10.18632/aging.202786] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022]
Abstract
Reticulophagy regulator 1 (RETEG1, also known as FAM134B) plays a crucial role in endoplasmic reticulum autophagy. We aimed to explore the effect of FAM134B-mediated endoplasmic reticulum autophagy in sepsis myocardial injury in mice. Sepsis myocardial injury mice were established via cecal ligation and puncture procedures. The expression of FAM134B and LC3-II/I was determined using immunohistochemistry. Myocardial tissue morphological changes and apoptosis were examined using hematoxylin and eosin (H&E) staining and TUNEL analysis. The effects of FAM134B knockdown or overexpression on mice with sepsis myocardial injury were also studied. The levels of TNF-α, IL-6, IL-8, and IL-10 were evaluated using enzyme-linked immunosorbent assay (ELISA). Autophagy- and apoptosis-related protein expression was detected using western blotting. The effect of FAM134B on Lipopolysaccharide (LPS) -induced cardiomyocytes was also studied. The expression of FAM134B and LC3-II/I increased in sepsis mice and lipopolysaccharide (LPS)-treated cardiomyocytes. 3-Methyladenine (3-MA) significantly inhibited FAM134B and LC3-II/I expression and promoted myocardial injury, inflammation response, and cardiomyocyte apoptosis. The overexpression of FAM134B could minimize myocardial injury, inflammation, and apoptosis, whereas FAM134B knockdown showed opposite effects. FAM134B-mediated endoplasmic reticulum autophagy had a protective effect on sepsis myocardial injury in mice by reducing inflammation and tissue apoptosis, which may provide new insights for sepsis myocardial injury therapies.
Collapse
Affiliation(s)
- Tong Li
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Yongsheng Chen
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, PR China
| | - Yue Li
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Zhipeng Yao
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Wenhua Liu
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| |
Collapse
|
10
|
Kabaklıoğlu M, Kaya M, Şahin IE, Gamsızkan M, Bahçıvan A, Eröz R. Short- and long-term effects of rapamycin on ischemic damage and apoptotic changes in torsion of rat testes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:85-94. [PMID: 32813042 DOI: 10.1007/s00210-020-01965-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
Rapamycin has antioxidant defense mechanisms and immune suppressive effects. To detect the short- and long-term effects of rapamycin on ischemic damage and apoptotic changes in torsion of rat testes, mature male albino Wistar rats (n = 48) were included in the study as control, sham, early torsion-detorsion (T/D), early rapamycin treatment, early rapamycin control, late T/D, late rapamycin treatment, and late rapamycin control. The right testis was rotated 720° in a clockwise direction during 4 h in operation groups. Rapamycin was administered orally three times: 30 min before detorsion and 24 and 48 h after detorsion. The animals were killed on the third day in early groups and on the tenth day in late groups after detorsion. Statistically significant differences among all groups were detected for SOD and TBARS, mean seminiferous tubule diameter (MSTD) and Cosentino's histologic score (CHS), caspase 3, bax, average number of apoptotic cells per tubule (ANPCT), and percentage of apoptotic tubule (PAT) values. ANPCT values were 10% lower in the rapamycin treatment groups compared with the untreated T/D groups, and the PAT values were also approximately 1.3 times lower. Although short-term usage of rapamycin may reduce to the tubular injury caused by I/R conversely to apoptosis in the testicular tissue after testicular torsion, rapamycin may have the potential to increase the long-term apoptosis with/without testicular torsion and a subsequent regression in fertility.
Collapse
Affiliation(s)
- Murat Kabaklıoğlu
- Department of Pediatric Surgery, Duzce University Medical Faculty, Duzce, Turkey.
| | - Murat Kaya
- Department of Pediatric Surgery, Duzce University Medical Faculty, Duzce, Turkey
| | - Ibrahim Ethem Şahin
- Department of Medical Biochemistry, Duzce University Medical Faculty, Duzce, Turkey
| | - Mehmet Gamsızkan
- Department of Medical Pathology, Duzce University Medical Faculty, Duzce, Turkey
| | - Atike Bahçıvan
- Department of Medical Pathology, Duzce University Medical Faculty, Duzce, Turkey
| | - Recep Eröz
- Department of Medical Genetics, Duzce University Medical Faculty, Duzce, Turkey
| |
Collapse
|
11
|
Still Living Better through Chemistry: An Update on Caloric Restriction and Caloric Restriction Mimetics as Tools to Promote Health and Lifespan. Int J Mol Sci 2020; 21:ijms21239220. [PMID: 33287232 PMCID: PMC7729921 DOI: 10.3390/ijms21239220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR), the reduction of caloric intake without inducing malnutrition, is the most reproducible method of extending health and lifespan across numerous organisms, including humans. However, with nearly one-third of the world’s population overweight, it is obvious that caloric restriction approaches are difficult for individuals to achieve. Therefore, identifying compounds that mimic CR is desirable to promote longer, healthier lifespans without the rigors of restricting diet. Many compounds, such as rapamycin (and its derivatives), metformin, or other naturally occurring products in our diets (nutraceuticals), induce CR-like states in laboratory models. An alternative to CR is the removal of specific elements (such as individual amino acids) from the diet. Despite our increasing knowledge of the multitude of CR approaches and CR mimetics, the extent to which these strategies overlap mechanistically remains unclear. Here we provide an update of CR and CR mimetic research, summarizing mechanisms by which these strategies influence genome function required to treat age-related pathologies and identify the molecular fountain of youth.
Collapse
|
12
|
Gong C, Xia H. Resveratrol suppresses melanoma growth by promoting autophagy through inhibiting the PI3K/AKT/mTOR signaling pathway. Exp Ther Med 2020; 19:1878-1886. [PMID: 32104244 PMCID: PMC7027143 DOI: 10.3892/etm.2019.8359] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
Resveratrol (RV) is a natural polyphenolic phytoalexin derived from peanuts, red grape skins and red wine, and has been demonstrated to alleviate multiple types of malignancies. However, how RV achieves this in melanoma is unknown. The aim of present study was to investigate the role of RV in melanoma, using Cell Counting Kit-8, flow cytometry and western blot analysis. RV inhibited melanoma cell viability, migration and invasion counteracting melanoma progression. In addition, proteins associated with autophagy, including Beclin 1 and microtubule-associated protein 1A/1B-light chain 3 (LC3)-II/I, were upregulated, whereas p62 expression was downregulated in RV-treated cells. The number of LC3+ puncta, which can be applied to represent autophagosome formation, increased following RV treatment, suggesting that RV may trigger autophagy in melanoma cells. Treatment with the autophagy inhibitor, 3-methyladenine, reversed the RV-dependent inhibition of viability, migration and invasion of melanoma cells. RV treatment also reduced the ratios of phosphorylated (p)-AKT/AKT and p-mTOR/mTOR in melanoma cells. In conclusion, these findings suggested that RV may inhibit the viability and migration of melanoma cells through inhibiting the AKT/mTOR pathway, thus triggering autophagy. This indicated that RV may serve as an innovative therapeutic for melanoma treatment.
Collapse
Affiliation(s)
- Changhua Gong
- Department of Pharmacy, People's Hospital of Zhenhai, Ningbo, Zhejiang 315202, P.R. China
| | - Honglei Xia
- Department of Pharmacy, People's Hospital of Zhenhai, Ningbo, Zhejiang 315202, P.R. China
| |
Collapse
|
13
|
Liu S, Yang Y, Gao H, Zhou N, Wang P, Zhang Y, Zhang A, Jia Z, Huang S. Trehalose attenuates renal ischemia-reperfusion injury by enhancing autophagy and inhibiting oxidative stress and inflammation. Am J Physiol Renal Physiol 2020; 318:F994-F1005. [PMID: 32068461 DOI: 10.1152/ajprenal.00568.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Renal ischemia-reperfusion (IR) injury is one of the most common acute kidney injuries, but there is still a lack of effective treatment in the clinical setting. Trehalose (Tre), a natural disaccharide, has been demonstrated to protect against oxidative stress, inflammation, and apoptosis. However, whether it could protect against IR-induced renal injury needs to be investigated. In an in vivo experiment, C57BL/6J mice were pretreated with or without Tre (2 g/kg) through a daily single intraperitoneal injection from 3 days before renal IR surgery. Renal function, apoptosis, oxidative stress, and inflammation were analyzed to evaluate kidney injury. In an in vitro experiment, mouse proximal tubular cells were treated with or without Tre under a hypoxia/reoxygenation condition. Western blot analysis, autophagy flux detection, and apoptosis assay were performed to evaluate the level of autophagy and antiapoptotic effect of Tre. The in vivo results showed that the renal damage induced by IR was ameliorated by Tre treatment, as renal histology and renal function were improved and the enhanced protein levels of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin were blocked. Moreover, autophagy was activated by Tre pretreatment along with inhibition of the IR injury-induced apoptosis, oxidative stress, and inflammation. The in vitro results showed that Tre treatment activated autophagy and protected against hypoxia/reoxygenation-induced tubular cell apoptosis and oxidative stress. Our results demonstrated that Tre protects against IR-induced renal injury, possibly by enhancing autophagy and blocking oxidative stress, inflammation, and apoptosis, suggesting its potential use for the clinical treatment of renal IR injury.
Collapse
Affiliation(s)
- Suwen Liu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yunwen Yang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Huiping Gao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Ning Zhou
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Peipei Wang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Fang J, Wang J, Chen F, Xu Y, Zhang H, Wang Y. α7nAChR Deletion Aggravates Myocardial Infarction and Enhances Systemic Inflammatory Reaction via mTOR-Signaling-Related Autophagy. Inflammation 2020; 42:1190-1202. [PMID: 30806956 DOI: 10.1007/s10753-019-00979-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alpha7 nicotinic acetylcholine receptor (α7nAChR) has been previously reported to play an alleviative role in myocardial infarction (MI). In this study, we investigated its specific mechanism. α7nAChR-/- mice and its control (α7nAChR+/+) were used for the study of α7nAChR. Left anterior descending coronary artery occlusion was conducted for the creation of mice MI model and lipopolysaccharide (LPS) was used as inflammatory stressor in murine peritoneal macrophages. Triphenyltetrazolium chloride (TTC) staining and echocardiography was used for the detection of infarct size and cardiac function, respectively. Western blot was conducted for the testing of autophagy-related proteins and enzyme-linked immunosorbent assay (ELISA) and real-time polymerase chain reaction (RT-PCR) was used for the testing of proinflammatory cytokines. Rapamycin was used for the induction of autophagy through inhibiting mammalian target of rapamycin (mTOR)-related signaling. We found that knocking out α7nAChR enhanced the cardiac infarct size and damaged cardiac function in MI. α7nAChR deficiency increased the levels of several proinflammatory cytokines in serum and spleen from MI mice as well as murine macrophages under inflammatory stress. α7nAChR deletion decreased the level of autophagy in spleen from MI mice and macrophages under inflammatory stress. Rapamycin alleviated the cardiac function and systemic inflammatory reaction in MI mice as well as inflammatory reaction in macrophages under inflammatory stress, which was attenuated by knocking out α7nAChR. Our current study investigated the mechanism of α7nAChR-mediated cardio-protective and anti-inflammatory effect related to mTOR-related autophagy, which might provide a novel insight in the treatment of MI.
Collapse
Affiliation(s)
- Jinyan Fang
- Department of Emergency, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 HuanSha Road, Hangzhou, Zhejiang, China
| | - Jiawei Wang
- Department of Endocrinology, The 903th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Fanghui Chen
- Department of Emergency, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 HuanSha Road, Hangzhou, Zhejiang, China
| | - Yuansheng Xu
- Department of Emergency, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 HuanSha Road, Hangzhou, Zhejiang, China
| | - Hao Zhang
- Department of Emergency, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 HuanSha Road, Hangzhou, Zhejiang, China
| | - Yi Wang
- Department of Emergency, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 HuanSha Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Zhao Y, Wang Z, Zhang W, Zhang L. MicroRNAs play an essential role in autophagy regulation in various disease phenotypes. Biofactors 2019; 45:844-856. [PMID: 31418958 PMCID: PMC6916288 DOI: 10.1002/biof.1555] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Autophagy is a highly conserved catabolic process and fundamental biological process in eukaryotic cells. It recycles intracellular components to provide nutrients during starvation and maintains quality control of organelles and proteins. In addition, autophagy is a well-organized homeostatic cellular process that is responsible for the removal of damaged organelles and intracellular pathogens. Moreover, it also modulates the innate and adaptive immune systems. Micro ribonucleic acids (microRNAs) are a mature class of post-transcriptional modulators that are widely expressed in tissues and organs. And, it can suppress gene expression by targeting messenger RNAs for translational repression or, at a lesser extent, degradation. Research indicates that microRNAs regulate autophagy through different pathways, playing an essential role in the treatment of various diseases. It is an important regulator of fundamental cellular processes such as proliferation, autophagy, and cell apoptosis. In this review article, we first review the current knowledge of autophagy and the function of microRNAs. Then, we summarize the mechanism of autophagy and the signaling pathways related to autophagy by citing at least the main proteins involved in the different phases of the process. Second, we introduce other members of RNA and report some examples in various pathologies. Finally, we review the current literature regarding microRNA-based therapies for cancer, atherosclerosis, cardiac disease, tuberculosis, and viral diseases. MicroRNAs can cause autophagy upregulation or downregulation by targeting genes or affecting autophagy-related signaling pathways. Therefore, the microRNAs have a huge potential in autophagy regulation, and it is the function as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Yunyi Zhao
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
| | - Ze Wang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
| | - Wenhui Zhang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
- Ministry of Education, Engineering Research Center for Bioreactor and Pharmaceutical DevelopmentJilin Agricultural UniversityChangchunChina
| | - Linbo Zhang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
- Ministry of Education, Engineering Research Center for Bioreactor and Pharmaceutical DevelopmentJilin Agricultural UniversityChangchunChina
| |
Collapse
|
16
|
Wang W, Sun W, Cheng Y, Xu Z, Cai L. Role of sirtuin-1 in diabetic nephropathy. J Mol Med (Berl) 2019; 97:291-309. [PMID: 30707256 PMCID: PMC6394539 DOI: 10.1007/s00109-019-01743-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/29/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Diabetic nephropathy (DN) is a research priority for scientists around the world because of its high prevalence and poor prognosis. Although several mechanisms have been shown to be involved in its pathogenesis and many useful drugs have been developed, the management of DN remains challenging. Increasing amounts of evidence show that silent information regulator 2 homolog 1 (sirtuin-1), a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, plays a crucial role in the pathogenesis and development of DN. Clinical data show that gene polymorphisms of sirtuin-1 affect patient vulnerability to DN. In addition, upregulation of sirtuin-1 attenuates DN in various experimental models of diabetes and in renal cells, including podocytes, mesangial cells, and renal proximal tubular cells, incubated with high concentrations of glucose or advanced glycation end products. Mechanistically, sirtuin-1 has its renoprotective effects by modulating metabolic homeostasis and autophagy, resisting apoptosis and oxidative stress, and inhibiting inflammation through deacetylation of histones and the transcription factors p53, forkhead box group O, nuclear factor-κB, hypoxia-inducible factor-1α, and others. Furthermore, some microRNAs have been implicated in the progression of DN because they target sirtuin-1 mRNA. Several synthetic drugs and natural compounds have been identified that upregulate the expression and activity of sirtuin-1, which protects against DN. The present review will summarize advances in knowledge regarding the role of sirtuin-1 in the pathogenesis of DN. The available evidence implies that sirtuin-1 has great potential as a clinical target for the prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Wanning Wang
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
- Pediatric Research Institute, Department of Pediatrics, The University of Louisville School of Medicine, Louisville, KY 40292 USA
| | - Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
| | - Yanli Cheng
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, The University of Louisville School of Medicine, Louisville, KY 40292 USA
- Departments of Radiation Oncology, Pharmacology and Toxicology, The University of Louisville School of Medicine, 570 S. Preston Str., Baxter I, Suite 304F, Louisville, KY 40292 USA
| |
Collapse
|
17
|
Ji J, Zhou X, Xu P, Li Y, Shi H, Chen D, Li R, Shi H. Deficiency of apoptosis-stimulating protein two of p53 ameliorates acute kidney injury induced by ischemia reperfusion in mice through upregulation of autophagy. J Cell Mol Med 2019; 23:2457-2467. [PMID: 30675758 PMCID: PMC6433670 DOI: 10.1111/jcmm.14094] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/04/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) has become a common disorder with a high risk of morbidity and mortality, which remains major medical problem without reliable and effective therapeutic intervention. Apoptosis-stimulating protein two of p53 (ASPP2) is a proapoptotic member that belongs to p53 binding protein family, which plays a key role in regulating apoptosis and cell growth. However, the role of ASPP2 in AKI has not been reported. To explore the role of ASPP2 in the progression of AKI, we prepared an AKI mouse model induced by ischaemia reperfusion (I/R) in wild-type (ASPP2+/+ ) mice and ASPP2 haploinsufficient (ASPP2+/- ) mice. The expression profile of ASPP2 were examined in wild-type mice. The renal injury, inflammation response, cellular apoptosis and autophagic pathway was assessed in ASPP2+/+ and ASPP2+/- mice. The renal injury, inflammation response and cellular apoptosis was analysed in ASPP2+/+ and ASPP2+/- mice treated with 3-methyladenine or vehicle. The expression profile of ASPP2 showed an increase at the early stage while a decrease at the late stage during renal injury. Compared with ASPP2+/+ mice, ASPP2 deficiency protected mice against renal injury induced by I/R, which mainly exhibited in slighter histologic changes, lower levels of blood urea nitrogen and serum creatinine, and less apoptosis as well as inflammatory response. Furthermore, ASPP2 deficiency enhanced autophagic activity reflecting in the light chain 3-II conversion and p62 degradation, while the inhibition of autophagy reversed the protective effect of ASPP2 deficiency on AKI. These data suggest that downregulation of ASPP2 can ameliorate AKI induced by I/R through activating autophagy, which may provide a novel therapeutic strage for AKI.
Collapse
Affiliation(s)
- Jing Ji
- Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Hepatology, Capital Medical University, Beijing, China.,Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xiaoshuang Zhou
- Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Ping Xu
- Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Yafeng Li
- Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Honglin Shi
- Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Dexi Chen
- Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Rongshan Li
- Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Hongbo Shi
- Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| |
Collapse
|