1
|
Xu TT, Deng YY, Yu XY, Li M, Fu YY. Natural autophagy modulators in non-communicable diseases: from autophagy mechanisms to therapeutic potential. Acta Pharmacol Sin 2024:10.1038/s41401-024-01356-y. [PMID: 39090393 DOI: 10.1038/s41401-024-01356-y] [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: 04/17/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
Non-communicable diseases (NCDs) are defined as a kind of diseases closely related to bad behaviors and lifestyles, e.g., cardiovascular diseases, cancer, and diabetes. Driven by population growth and aging, NCDs have become the biggest disease burden in the world, and it is urgent to prevent and control these chronic diseases. Autophagy is an evolutionarily conserved process that degrade cellular senescent or malfunctioning organelles in lysosomes. Mounting evidence has demonstrated a major role of autophagy in the pathogenesis of cardiovascular diseases, cancer, and other major human diseases, suggesting that autophagy could be a candidate therapeutic target for NCDs. Natural products/phytochemicals are important resources for drugs against a wide variety of diseases. Recently, compounds from natural plants, such as resveratrol, curcumin, and ursolic acid, have been recognized as promising autophagy modulators. In this review, we address recent advances and the current status of the development of natural autophagy modulators in NCDs and provide an update of the latest in vitro and in vivo experiments that pave the way to clinical studies. Specifically, we focus on the relationship between natural autophagy modulators and NCDs, with an intent to identify natural autophagy modulators with therapeutic potential.
Collapse
Affiliation(s)
- Ting-Ting Xu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ying-Yi Deng
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xi-Yong Yu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Min Li
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Yuan-Yuan Fu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
2
|
Ponticelli C, Reggiani F, Moroni G. Autophagy: A Silent Protagonist in Kidney Transplantation. Transplantation 2024; 108:1532-1541. [PMID: 37953477 DOI: 10.1097/tp.0000000000004862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Autophagy is a lysosome-dependent regulated mechanism that recycles unnecessary cytoplasmic components. It is now known that autophagy dysfunction may have a pathogenic role in several human diseases and conditions, including kidney transplantation. Both defective and excessive autophagy may induce or aggravate several complications of kidney transplantation, such as ischemia-reperfusion injury, alloimmune response, and immunosuppressive treatment and side effects. Although it is still complicated to measure autophagy levels in clinical practice, more attention should be paid to the factors that may influence autophagy. In kidney transplantation, the association of low doses of a mammalian target of rapamycin inhibitor with low doses of a calcineurin inhibitor may be of benefit for autophagy modulation. However, further studies are needed to explore the role of other autophagy regulators.
Collapse
Affiliation(s)
| | - Francesco Reggiani
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gabriella Moroni
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
3
|
Liu Y, Mou L, Yi Z, Lin Q, Banu K, Wei C, Yu X. Integrative informatics analysis identifies that ginsenoside Re improves renal fibrosis through regulation of autophagy. J Nat Med 2024; 78:722-731. [PMID: 38683298 DOI: 10.1007/s11418-024-01800-7] [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: 07/19/2023] [Accepted: 03/01/2024] [Indexed: 05/01/2024]
Abstract
We previously demonstrated that ginsenoside Re (G-Re) has protective effects on acute kidney injury. However, the underlying mechanism is still unclear. In this study, we conducted a meta-analysis and pathway enrichment analysis of all published transcriptome data to identify differentially expressed genes (DEGs) and pathways of G-Re treatment. We then performed in vitro studies to measure the identified autophagy and fibrosis markers in HK2 cells. In vivo studies were conducted using ureteric obstruction (UUO) and aristolochic acid nephropathy (AAN) models to evaluate the effects of G-Re on autophagy and kidney fibrosis. Our informatics analysis identified autophagy-related pathways enriched for G-Re treatment. Treatment with G-Re in HK2 cells reduced autophagy and mRNA levels of profibrosis markers with TGF-β stimulation. In addition, induction of autophagy with PP242 neutralized the anti-fibrotic effects of G-Re. In murine models with UUO and AAN, treatment with G-Re significantly improved renal function and reduced the upregulation of autophagy and profibrotic markers. A combination of informatics analysis and biological experiments confirmed that ginsenoside Re could improve renal fibrosis and kidney function through the regulation of autophagy. These findings provide important insights into the mechanisms of G-Re's protective effects in kidney injuries.
Collapse
Affiliation(s)
- Yingying Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lingyun Mou
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute of Biochemistry and Molecular Biology, School of Life Science, Lanzhou University, Lanzhou, China
| | - Zhengzi Yi
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qisheng Lin
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Khadija Banu
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chengguo Wei
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Xiaoxia Yu
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Division of Nephrology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.
| |
Collapse
|
4
|
Woon TH, Tan MJH, Kwan YH, Fong W. Evidence of the interactions between immunosuppressive drugs used in autoimmune rheumatic diseases and Chinese herbal medicine: A scoping review. Complement Ther Med 2024; 80:103017. [PMID: 38218549 DOI: 10.1016/j.ctim.2024.103017] [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: 09/24/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
OBJECTIVES Chinese herbal medicine (CHM) has been shown to be effective in autoimmune rheumatic diseases, but harmful herb-drug interactions might be inherent. We aim to review the evidence regarding herb-drug interactions between immunosuppressive drugs used in autoimmune rheumatic diseases and CHM. METHODS We searched PubMed, EMBASE and CINAHL from inception till 30 April 2023 using keywords that encompassed 'herb-drug interactions', 'herbs' and 'immunosuppressants'. Articles were included if they contained reports about interactions between immunosuppressive drugs used in the treatment of rheumatic diseases with CHM. Level of evidence for each pair of interaction was graded using the algorithm developed by Colalto. RESULTS A total of 65 articles and 44 unique pairs of interactions were identified. HDIs were reported for cyclophosphamide, cyclosporine, tacrolimus, methotrexate, mycophenolic acid, glucocorticoids, sulfasalazine, tofacitinib and biologic disease-modifying antirheumatic drugs. Among these, cyclosporine (n = 27, 41.5%) and tacrolimus (n = 19, 29.2%) had the highest number of documented interactions. Hypericum perforatum had the highest level of evidence of interaction with cyclosporine and tacrolimus. Consumption reduced the bioavailability and therapeutic effects of the drugs. Schisandra sphenanthera had the highest level of evidence of interaction with tacrolimus and increased the bioavailability of the drug. Majority of the articles were animal studies. CONCLUSION Overall level of evidence for the included studies were low, though interactions between cyclosporine, tacrolimus, Hypericum perforatum and Schisandra sphenanthera were the most and well-documented. Healthcare professionals should actively enquire about the concurrent use of CHM in patients, especially when drugs with a narrow therapeutic index are consumed.
Collapse
Affiliation(s)
- Ting Hui Woon
- Department of Rheumatology and Immunology, Singapore General Hospital, 20 College Road, Singapore 169856, Singapore
| | - Melissa Jia Hui Tan
- Department of Pharmacy, Sengkang General Hospital, 110 Sengkang E Way, Singapore 544886, Singapore
| | - Yu Heng Kwan
- Department of Rheumatology and Immunology, Singapore General Hospital, 20 College Road, Singapore 169856, Singapore; Program in Health Services and Systems Research, Duke-NUS Medical School, 8 College Rd, Singapore 169857, Singapore; Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Singapore
| | - Warren Fong
- Department of Rheumatology and Immunology, Singapore General Hospital, 20 College Road, Singapore 169856, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore; Office of Education, Duke-NUS Medical School, 8 College Rd, Singapore 169857, Singapore.
| |
Collapse
|
5
|
Vidigal AC, de Lucena DD, Beyerstedt S, Rangel ÉB. A comprehensive update of the metabolic and toxicological considerations for immunosuppressive drugs used during pancreas transplantation. Expert Opin Drug Metab Toxicol 2023; 19:405-427. [PMID: 37542452 DOI: 10.1080/17425255.2023.2243808] [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: 03/26/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Despite significant advancements in immunosuppressive regimens and surgical techniques, the prevalence of adverse events related to immunosuppression remains a major challenge affecting the long-term survival rates of pancreas and kidney allografts. AREAS COVERED This article presents a comprehensive review of the literature and knowledge (Jan/2012-Feb/2023) concerning glucose metabolism disorders and nephrotoxicity associated with tacrolimus and mammalian target of rapamycin inhibitors (mTORi). Novel signaling pathways potentially implicated in these adverse events are discussed. Furthermore, we extensively examine the findings from clinical trials evaluating the efficacy and safety of tacrolimus, mTORi, and steroid minimization. EXPERT OPINION Tacrolimus-based regimens continue to be the standard treatment following pancreas transplants. However, prolonged use of tacrolimus and mTORi may lead to hyperglycemia and nephrotoxicity. Understanding and interpreting experimental data, particularly concerning novel signaling pathways beyond calcineurin-NFAT and mTOR pathways, can offer valuable insights for therapeutic interventions to mitigate hyperglycemia and nephrotoxicity. Additionally, critically analyzing clinical trial results can identify opportunities for personalized safety-based approaches to minimize side effects. It is imperative to conduct randomized-controlled studies to assess the impact of mTORi use and steroid-free protocols on pancreatic allograft survival. Such studies will aid in tailoring treatment strategies for improved transplant outcomes.
Collapse
Affiliation(s)
- Ana Cláudia Vidigal
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
| | - Débora D de Lucena
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, São Paulo, Brazil
| | - Érika B Rangel
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, São Paulo, Brazil
| |
Collapse
|
6
|
Nemati M, Ebrahimi Z, Karbalaei N, Dastghaib S, Khakshournia S, Sargazi M. In Vitro and In Vivo Improvement of Islet Quality and Transplantation Successes following Islet Treatment with Biomaterials in Diabetic Rats. J Diabetes Res 2023; 2023:1399917. [PMID: 37265573 PMCID: PMC10232112 DOI: 10.1155/2023/1399917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/08/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023] Open
Abstract
Background Loss of islet survival and function, caused by native niche disruption and oxidative stress induction during mechanical and enzymatic isolation, limits the effectiveness of islet transplantation. Reconstitution of islet microenvironment, vascularization, and decreased oxidative stress with biomaterials may improve islet quality and graft outcomes. We investigated effects of two biomaterials, platelet-rich plasma and pancreatic islets homogenate combination on islet recovery and quality by evaluating in vitro islet survival, secretory function, and oxidative stress parameters and assessing in vivo transplantation outcomes. Methods In vitro, islet viability and secretory function of isolated islets were assessed after 24 h and 72 h incubation with biomaterials. Also, oxidative stress markers were measured once after isolation and 24 h after incubation with biomaterials. For evaluating in vivo effects, cultured islets for 24 h were transplanted into subscapular space of diabetic rat kidney, and outcomes were analyzed by measuring serum glucose and insulin concentrations, glucose tolerance test, level of oxidative parameters, and pancreatic gene expression. Results Treating islets with biomaterials significantly increased their viability and secretory function, reduced MDA level, and elevate SOD and CAT activity. Decreased level of glucose and MDA improved insulin level, increased SOD activity, and also enhanced pdx1 and insulin gene expression in diabetic rats after islet transplantation. Conclusions Biomaterials used in the present study should be consider as beneficial materials for increasing islet transplantation outcome. These materials may hamper transplantation limitation to some extent.
Collapse
Affiliation(s)
- Marzieh Nemati
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ebrahimi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Narges Karbalaei
- Department of physiology, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Authophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Khakshournia
- Department of Biochemistry, Shiraz University of Medical Science, Shiraz, Iran
| | - Mojtaba Sargazi
- Department of physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
How ginseng regulates autophagy: Insights from multistep process. Biomed Pharmacother 2023; 158:114139. [PMID: 36580724 DOI: 10.1016/j.biopha.2022.114139] [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: 10/20/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Although autophagy is a recognized contributor to the pathogenesis of human diseases, chloroquine and hydroxychloroquine are the only two FDA-approved autophagy inhibitors to date. Emerging evidence has revealed the potential therapeutic benefits of various extracts and active compounds isolated from ginseng, especially ginsenosides and their derivatives, by mediating autophagy. Mechanistically, active components from ginseng mediate key regulators in the multistep processes of autophagy, namely, initiation, autophagosome biogenesis and cargo degradation. AIM OF REVIEW To date, a review that systematically described the relationship between ginseng and autophagy is still lacking. Breakthroughs in finding the key players in ginseng-autophagy regulation will be a promising research area, and will provide positive insights into the development of new drugs based on ginseng and autophagy. KEY SCIENTIFIC CONCEPTS OF REVIEW Here, we comprehensively summarized the critical roles of ginseng-regulated autophagy in treating diseases, including cancers, neurological disorders, cardiovascular diseases, inflammation, and neurotoxicity. The dual effects of the autophagy response in certain diseases are worthy of note; thus, we highlight the complex impacts of both ginseng-induced and ginseng-inhibited autophagy. Moreover, autophagy and apoptosis are controlled by multiple common upstream signals, cross-regulate each other and affect certain diseases, especially cancers. Therefore, this review also discusses the cross-signal transduction pathways underlying the molecular mechanisms and interaction between ginseng-regulated autophagy and apoptosis.
Collapse
|
8
|
Yang K, Cao F, Wang W, Tian Z, Yang L. The relationship between HMGB1 and autophagy in the pathogenesis of diabetes and its complications. Front Endocrinol (Lausanne) 2023; 14:1141516. [PMID: 37065747 PMCID: PMC10090453 DOI: 10.3389/fendo.2023.1141516] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by elevated blood glucose levels and has become the third leading threat to human health after cancer and cardiovascular disease. Recent studies have shown that autophagy is closely associated with diabetes. Under normal physiological conditions, autophagy promotes cellular homeostasis, reduces damage to healthy tissues and has bidirectional effects on regulating diabetes. However, under pathological conditions, unregulated autophagy activation leads to cell death and may contribute to the progression of diabetes. Therefore, restoring normal autophagy may be a key strategy to treat diabetes. High-mobility group box 1 protein (HMGB1) is a chromatin protein that is mainly present in the nucleus and can be actively secreted or passively released from necrotic, apoptotic, and inflammatory cells. HMGB1 can induce autophagy by activating various pathways. Studies have shown that HMGB1 plays an important role in insulin resistance and diabetes. In this review, we will introduce the biological and structural characteristics of HMGB1 and summarize the existing knowledge on the relationship between HMGB1, autophagy, diabetes, and diabetic complications. We will also summarize potential therapeutic strategies that may be useful for the prevention and treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Kun Yang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Cao
- College of Acupuncture and Massage, Beijing University of Chinese Medicine, Beijing, China
- Department of Acupuncture, Haidian District Shuangyushu Community Health Service Center, Beijing, China
| | - Weili Wang
- Institute of Basic Research in Clinical Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyu Tian
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Lu Yang, ; Zhenyu Tian,
| | - Lu Yang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lu Yang, ; Zhenyu Tian,
| |
Collapse
|
9
|
Miedziaszczyk M, Bajon A, Jakielska E, Primke M, Sikora J, Skowrońska D, Idasiak-Piechocka I. Controversial Interactions of Tacrolimus with Dietary Supplements, Herbs and Food. Pharmaceutics 2022; 14:pharmaceutics14102154. [PMID: 36297591 PMCID: PMC9611668 DOI: 10.3390/pharmaceutics14102154] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 12/05/2022] Open
Abstract
Tacrolimus is an immunosuppressive calcineurin inhibitor used to prevent rejection in allogeneic organ transplant recipients, such as kidney, liver, heart or lung. It is metabolized in the liver, involving the cytochrome P450 (CYP3A4) isoform CYP3A4, and is characterized by a narrow therapeutic window, dose-dependent toxicity and high inter-individual and intra-individual variability. In view of the abovementioned facts, the aim of the study is to present selected interactions between tacrolimus and the commonly used dietary supplements, herbs and food. The review was based on the available scientific literature found in the PubMed, Scopus and Cochrane databases. An increase in the serum concentration of tacrolimus can be caused by CYP3A4 inhibitors, such as grapefruit, pomelo, clementine, pomegranate, ginger and turmeric, revealing the side effects of this drug, particularly nephrotoxicity. In contrast, CYP3A4 inducers, such as St. John’s Wort, may result in a lack of therapeutic effect by reducing the drug concentration. Additionally, the use of Panax ginseng, green tea, Schisandra sphenanthera and melatonin in patients receiving tacrolimus is highly controversial. Therefore, since alternative medicine constitutes an attractive treatment option for patients, modern healthcare should emphasize the potential interactions between herbal medicines and synthetic drugs. In fact, each drug or herbal supplement should be reported by the patient to the physician (concordance) if it is taken in the course of immunosuppressive therapy, since it may affect the pharmacokinetic and pharmacodynamic parameters of other preparations.
Collapse
Affiliation(s)
- Miłosz Miedziaszczyk
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
- Correspondence:
| | - Aleksander Bajon
- Student’s Scientific Section of Nephrology and Clinical Transplantology, 61-701 Poznan, Poland
| | - Ewelina Jakielska
- Student’s Scientific Section of Nephrology and Clinical Transplantology, 61-701 Poznan, Poland
| | - Marta Primke
- Student’s Scientific Section of Nephrology and Clinical Transplantology, 61-701 Poznan, Poland
| | - Jędrzej Sikora
- Student’s Scientific Section of Nephrology and Clinical Transplantology, 61-701 Poznan, Poland
| | - Dagmara Skowrońska
- Student’s Scientific Section of Nephrology and Clinical Transplantology, 61-701 Poznan, Poland
| | - Ilona Idasiak-Piechocka
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
10
|
Effect of histone deacetylase inhibitor (vorinostat) on new-onset diabetes induced by tacrolimus. J Taibah Univ Med Sci 2022; 18:9-18. [DOI: 10.1016/j.jtumed.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
|
11
|
Eguchi N, Damyar K, Alexander M, Dafoe D, Lakey JRT, Ichii H. Anti-Oxidative Therapy in Islet Cell Transplantation. Antioxidants (Basel) 2022; 11:1038. [PMID: 35739935 PMCID: PMC9219662 DOI: 10.3390/antiox11061038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 01/27/2023] Open
Abstract
Islet cell transplantation has become a favorable therapeutic approach in the treatment of Type 1 Diabetes due to the lower surgical risks and potential complications compared to conventional pancreas transplantation. Despite significant improvements in islet cell transplantation outcomes, several limitations hamper long-term graft survival due to tremendous damage and loss of islet cells during the islet cell transplantation process. Oxidative stress has been identified as an omnipresent stressor that negatively affects both the viability and function of isolated islets. Furthermore, it has been established that at baseline, pancreatic β cells exhibit reduced antioxidative capacity, rendering them even more susceptible to oxidative stress during metabolic stress. Thus, identifying antioxidants capable of conferring protection against oxidative stressors present throughout the islet transplantation process is a valuable approach to improving the overall outcomes of islet cell transplantation. In this review we discuss the potential application of antioxidative therapy during each step of islet cell transplantation.
Collapse
Affiliation(s)
- Natsuki Eguchi
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Kimia Damyar
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Michael Alexander
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Donald Dafoe
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Jonathan R. T. Lakey
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92686, USA
| | - Hirohito Ichii
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| |
Collapse
|
12
|
Jeon H, Kim HY, Bae CH, Lee Y, Koo S, Kim S. Korean red ginseng decreases 1-methyl-4-phenylpyridinium-induced mitophagy in SH-SY5Y cells. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:537-544. [PMID: 34580047 DOI: 10.1016/j.joim.2021.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 07/19/2021] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Mitophagy is known to contribute towards progression of Parkinson's disease. Korean red ginseng (KRG) is a widely used medicinal herb in East Asia, and recent studies have reported that KRG prevents 1-methyl-4-phenylpyridinium ion (MPP+)-induced cell death. This study was undertaken to investigate whether KRG suppresses MPP+-induced apoptosis and mitophagy. METHODS SH-SY5Y cells were incubated with KRG for 24 h, and subsequently exposed to MPP+. The MPP+-induced cell death was confirmed with the 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, and the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay. Changes in the structure and function of mitochondria were confirmed using mitotracker, MitoSOX red mitochondrial superoxide indicator, parkin, and phosphatase and tensin homolog deleted on chromosome ten-induced putative kinase 1 (PINK1) immunofluorescent staining. Western blotting was performed to evaluate the expression of apoptosis-related factors in whole cells, including Bax, Bcl-2 and cleaved caspase-3, and mitophagy-related factors in the mitochondrial fraction, including cytochrome c, parkin, PINK1, translocase of the outer membrane 20 (TOM20), p62 and Beclin 1. RESULTS MPP+ induced cell death by cytochrome c release and caspase-3 activation; however, this effect was suppressed by KRG's regulation of the expressions of Bcl-2 and Bax. Moreover, MPP+ exposure increased the mitochondrial expressions of parkin, PINK1, Beclin 1 and p62, and decreased TOM20, cytochrome c and Bcl-2 expressions. These MPP+-induced changes in the mitochondrial fraction were attenuated by treatment with KRG. CONCLUSION KRG effectively prevents MPP+-induced SH-SY5Y cell death by regulating cytochrome c release from mitochondria and PINK1/parkin-mediated mitophagy, through regulation of the Bcl-2 family.
Collapse
Affiliation(s)
- Hyongjun Jeon
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea; Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Hee-Young Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Chang-Hwan Bae
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea; Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Yukyung Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea; Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Sungtae Koo
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea; Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Seungtae Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea; Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea.
| |
Collapse
|
13
|
Vitamin E and ginseng combined supplement for treatment of male erectile dysfunction: A double-blind, placebo-controlled, randomized, clinical trial. ADVANCES IN INTEGRATIVE MEDICINE 2021. [DOI: 10.1016/j.aimed.2019.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
14
|
Quan Y, Luo K, Cui S, Lim SW, Shin YJ, Ko EJ, Kim JH, Chung SJ, Bae SK, Chung BH, Yang CW. The therapeutic efficacy of water-soluble coenzyme Q10 in an experimental model of tacrolimus-induced diabetes mellitus. Korean J Intern Med 2020; 35:1443-1456. [PMID: 32279476 PMCID: PMC7652663 DOI: 10.3904/kjim.2019.269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/01/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIMS Coenzyme Q10 (CoQ10) has antioxidant effects and is commercially available and marketed extensively. However, due to its low bioavailability, its effects are still controversial. We developed a water-soluble CoQ10-based micelle formulation (CoQ10-W) and tested it in an experimental model of tacrolimus (TAC)-induced diabetes mellitus (DM). METHODS We developed CoQ10-W from a glycyrrhizic-carnitine mixed layer CoQ10 micelle preparation based on acyltransferases. TAC-induced DM rats were treated with either lipid-soluble CoQ10 (CoQ10-L) or CoQ10-W for 4 weeks. Their plasma and pancreatic CoQ10 concentrations were measured using liquid chromatography- tandem mass spectrometry. The therapeutic efficacies of CoQ10-W and CoQ10-L on TAC-induced DM were compared using functional and morphological parameters and their effects on cell viability and reactive oxygen species (ROS) production were also evaluated in cultured rat insulinoma cells. RESULTS The plasma CoQ10 level was significantly increased in the CoQ10-W group compared to that in the CoQ10-L group. Intraperitoneal glucose tolerance tests and glucose-stimulated insulin secretion revealed that CoQ10-W controlled hyperglycemia and restored insulin secretion significantly better than CoQ10-L. The TAC-mediated decrease in pancreatic islet size was significantly attenuated by CoQ10-W but not by CoQ10-L. TAC-induced oxidative stress and apoptosis were significantly more reduced by CoQ10-W than CoQ10-L. Electron microscopy revealed that CoQ10-W restored TAC-induced attenuation in the number of insulin granules and the average mitochondrial area, unlike CoQ10-L. In vitro studies showed that CoQ10-L and CoQ10-W both improved cell viability and reduced ROS production in TAC-treated islet cells to a similar extent. CONCLUSION CoQ10-W has better therapeutic efficacy than CoQ10-L in TAC-induced DM.
Collapse
Affiliation(s)
- Yi Quan
- Transplant Research Center, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
| | - Kang Luo
- Transplant Research Center, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
| | - Sheng Cui
- Transplant Research Center, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
| | - Sun Woo Lim
- Transplant Research Center, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
| | - Yoo Jin Shin
- Transplant Research Center, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
| | - Eun Jeong Ko
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Sang J. Chung
- Sungkyunkwan University School of Pharmacy, Suwon, Korea
| | - Soo Kyung Bae
- College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Transplant Research Center, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Correspondence to Chul Woo Yang, M,D Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, Korea Tel: +82-2-2258-6851 Fax: +82-2-2258-6917 E-mail:
| |
Collapse
|
15
|
Park SK, Hyun SH, In G, Park CK, Kwak YS, Jang YJ, Kim B, Kim JH, Han CK. The antioxidant activities of Korean Red Ginseng ( Panax ginseng) and ginsenosides: A systemic review through in vivo and clinical trials. J Ginseng Res 2020; 45:41-47. [PMID: 33437155 PMCID: PMC7790892 DOI: 10.1016/j.jgr.2020.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022] Open
Abstract
A wide range of studies have steadily pointed out the relation of oxidative stress to the primary and secondary causes of human disease and aging. As such, there have been multiple misconceptions about oxidative stress. Most of reactive oxygen species (ROS) generated from chronic diseases cause oxidative damage to cell membrane lipids and proteins. ROS production is increased by abnormal stimulation inside and outside in the body, and even though ROS are generated in cells in response to abnormal metabolic processes such as disease, it does not mean that they directly contribute to the pathogenesis of a disease. Therefore, the focus of treatment should not be on ROS production itself but on the prevention and treatment of diseases linked to ROS production, including types 1 and 2 diabetes, cancer, heart disease, schizophrenia, Parkinson's disease, and Alzheimer's disease. In this regard, Korean Red Ginseng (KRG) has been traditionally utilized to help prevent and treat diseases such as diabetes, cancer, inflammation, nervous system diseases, cardiovascular disease, and hyperlipidemia. Therefore, this review was intended to summarize in vivo animal and human clinical studies on the antioxidant activities of KRG and its components, ginsenosides.
Collapse
Affiliation(s)
- Soo Kyung Park
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Sun Hee Hyun
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Gyo In
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Chae-Kyu Park
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Yi-Seong Kwak
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Young-Jin Jang
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Chang-Kyun Han
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
16
|
Lim SW, Luo K, Quan Y, Cui S, Shin YJ, Ko EJ, Chung BH, Yang CW. The safety, immunological benefits, and efficacy of ginseng in organ transplantation. J Ginseng Res 2020; 44:399-404. [PMID: 32372861 PMCID: PMC7195583 DOI: 10.1016/j.jgr.2020.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/17/2020] [Accepted: 02/03/2020] [Indexed: 01/05/2023] Open
Abstract
Korean ginseng (Panax ginseng) is associated with a variety of therapeutic effects, including antioxidative, anti-inflammatory, vasorelaxative, antiallergic, antidiabetic, and anticancer effects. Accordingly, the use of ginseng has reached an all-time high among members of the general public. However, the safety and efficacy of ginseng in transplant recipients receiving immunosuppressant drugs have still not been elucidated. Transplantation is the most challenging and complex of surgical procedures and may require causation for the use of ginseng. In this regard, we have previously examined the safety, immunological benefits, and protective mechanisms of ginseng with respect to calcineurin inhibitor-based immunosuppression, which is the most widely used regimen in organ transplantation. Using an experimental model of calcineurin inhibitor-induced organ injury, we found that ginseng does not affect drug levels in the peripheral blood and tissue, favorably regulates immune response, and protects against calcineurin inhibitor-induced nephrotoxicity and pancreatic islet injury. On the basis of our experimental studies and a review of the related literature, we propose that ginseng may provide benefits in organ transplant recipients administered calcineurin inhibitors. Through the present review, we aimed to briefly discuss our current understanding of the therapeutic benefits of ginseng related to transplant patient survival.
Collapse
Affiliation(s)
- Sun Woo Lim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang Luo
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yi Quan
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sheng Cui
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Jin Shin
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Ko
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Nephrology Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Nephrology Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Nephrology Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
17
|
Role of Klotho in Chronic Calcineurin Inhibitor Nephropathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1825018. [PMID: 31772699 PMCID: PMC6854173 DOI: 10.1155/2019/1825018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 12/13/2022]
Abstract
Calcineurin inhibitors (CNIs) are the most popular immunosuppressants in organ transplantation, but nephrotoxicity is a major concern. The common mechanism underlying chronic CNI nephropathy is oxidative stress, and the process of chronic CNI nephropathy is similar to that of aging. Current studies provide evidence that antiaging Klotho protein plays an important role in protecting against oxidative stress, and its signaling is a target for preventing oxidative stress-induced aging process. In this review, we focus on the association between Klotho and oxidative stress and the protective mechanism of action of Klotho against oxidative stress in chronic CNI nephropathy. In addition, we discuss the delivery strategy for Klotho in CNI-induced nephropathy.
Collapse
|
18
|
Lim SW, Shin YJ, Luo K, Quan Y, Cui S, Ko EJ, Chung BH, Yang CW. Ginseng increases Klotho expression by FoxO3-mediated manganese superoxide dismutase in a mouse model of tacrolimus-induced renal injury. Aging (Albany NY) 2019; 11:5548-5569. [PMID: 31400753 PMCID: PMC6710054 DOI: 10.18632/aging.102137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/29/2019] [Indexed: 11/25/2022]
Abstract
The antioxidant function of Klotho is well-documented as a regulatory factor implicated in countering the aging process. This study investigated whether ginseng upregulates Klotho and its antiaging signaling in a setting of calcineurin inhibitor-induced oxidative stress. Although tacrolimus treatment reduced Klotho level in the serum and kidney, ginseng treatment was found to reverse the levels. Tacrolimus-induced oxidative stress was reduced by ginseng treatment, with functional and histological improvements. Effect of ginseng on Klotho-induced manganese superoxide dismutase signaling pathway during tacrolimus treatment in mice revealed that ginseng suppressed phosphatidylinositol 3-kinase/serine-threonine kinase Akt-mediated phosphorylation of forkhead box protein O3a and promoted the binding of forkhead box protein O3a to manganese superoxide dismutase promoter. In the mitochondria, ginseng reduced mitochondrial reactive oxygen species production, mitochondrial membrane potential, and oxygen consumption rate, whereas blocking phosphatidylinositol 3-kinase activity with LY294002 enhanced them. These findings together suggested that ginseng attenuated tacrolimus-induced oxidative stress via signaling between Klotho and the phosphatidylinositol 3-kinase/serine-threonine kinase Akt/forkhead box protein O3a-related antioxidant pathway.
Collapse
Affiliation(s)
- Sun Woo Lim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Jin Shin
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang Luo
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yi Quan
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sheng Cui
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Ko
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Transplant Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
19
|
Qomaladewi NP, Kim MY, Cho JY. Autophagy and its regulation by ginseng components. J Ginseng Res 2019; 43:349-353. [PMID: 31308805 PMCID: PMC6606841 DOI: 10.1016/j.jgr.2018.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 12/25/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023] Open
Abstract
Autophagy is the sequential process whereby cell components are degraded, which can occur due to nutrient deprivation. Its regulation has an essential role in many diseases, functioning in both cell survival and cell death. Autophagy starts when mTORC1 is inhibited, resulting in the activation of several complexes to form a cargo that fuses with a lysosome, where it undergoes degradation. In this review, we describe a plant extract that is well known in Korea, namely Korean ginseng extract; we studied how its derivatives and metabolites can regulate autophagy and thus mediate the pathogenesis of certain diseases.
Collapse
Affiliation(s)
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
20
|
Luo K, Lim SW, Jin J, Jin L, Gil HW, Im DS, Hwang HS, Yang CW. Cilastatin protects against tacrolimus-induced nephrotoxicity via anti-oxidative and anti-apoptotic properties. BMC Nephrol 2019; 20:221. [PMID: 31200653 PMCID: PMC6570925 DOI: 10.1186/s12882-019-1399-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 05/28/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Cilastatin (CL) is an inhibitor of dehydropeptidase-I, which is safely used in clinical practice to prevent nephrotoxicity of antibiotics. Tacrolimus (TAC) is the most important immunosuppressant in renal transplantation, but it causes considerable nephrotoxicity. We evaluated the protective effects of CL against chronic TAC-induced nephropathy. METHODS Chronic nephropathy was induced by administering TAC (1.5 mg/kg/ day, subcutaneous injection) to rats on a low-salt diet for 4 weeks. CL (75 or 150 mg/kg/day, intraperitoneal injection) was concomitantly treated with TAC. Human proximal tubular cells were exposed to TAC (50 μg/mL) with or without CL (250 μg/mL). We investigated the effects of CL on TAC-induced injury in terms of renal function, tubulointerstitial fibrosis, and inflammation. The effects of CL on oxidative stress and apoptosis were evaluated in both in vivo and in vitro models of TAC nephrotoxicity. RESULTS CL treatment improved TAC-induced renal dysfunction and decreased renal interstitial fibrosis (reduced expression of e-cadherin and TGFβ-1) and interstitial inflammation (decreased infiltration of ED-1-positive and osteopontin-positive cells). Compared to TAC treatment alone, CL co-treatment reduced oxidative stress (serum 8-OHdG level and immunoreactivity of 8-OHdG and 4-HHE in renal tissue) and increased renal expression of anti-oxidant enzyme, manganese superoxide dismutase. CL treatment decreased apoptotic cell death (decreased TUNEL-positive cells and reduced expression of active caspase-3) in TAC-treated kidney. In vitro CL treatment prevented tubular cell death from TAC treatment and decreased number of annexin V-positive cells were observed in cilastatin-cotreated cells. CONCLUSION CL has protective effects against chronic TAC-induced nephrotoxicity owing to its anti-oxidative and anti-apoptotic properties.
Collapse
Affiliation(s)
- Kang Luo
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Nephrology, Yanbian University Hospital, Yanbian, China
| | - Sun Woo Lim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jian Jin
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Nephrology, Yanbian University Hospital, Yanbian, China
| | - Long Jin
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyo Wook Gil
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, South Korea
| | - Dai Sig Im
- Department of Chemistry, College of Natural Sciences, Soonchunhyang University, Asan, South Korea.,SH Company, Asan, Chungnam, South Korea
| | - Hyeon Seok Hwang
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, South Korea.
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
21
|
Luo K, Yu JH, Quan Y, Shin YJ, Lee KE, Kim HL, Ko EJ, Chung BH, Lim SW, Yang CW. Therapeutic potential of coenzyme Q 10 in mitochondrial dysfunction during tacrolimus-induced beta cell injury. Sci Rep 2019; 9:7995. [PMID: 31142763 PMCID: PMC6541596 DOI: 10.1038/s41598-019-44475-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
We previously reported that oxidative stress induced by long-term tacrolimus treatment impairs mitochondrial function in pancreatic beta cells. In this study, we aimed to investigate the therapeutic potential of coenzyme Q10, which is known to be a powerful antioxidant, in mitochondrial dysfunction in tacrolimus-induced diabetic rats. In a rat model of tacrolimus-induced diabetes mellitus, coenzyme Q10 treatment improved pancreatic beta cell function. The administration of coenzyme Q10 improved insulin immunoreactivity within islets, which was accompanied by reductions in oxidative stress and apoptosis. Assessment of the mitochondrial ultrastructure by electron microscopy revealed that coenzyme Q10 treatment increased the size, number, and volume of mitochondria, as well as the number of insulin granules compared with that induced by tacrolimus treatment alone. An in vitro study using a pancreatic beta cell line showed that tacrolimus treatment increased apoptosis and the production of mitochondrial reactive oxygen species, while cotreatment with coenzyme Q10 effectively attenuated these alterations. At the subcellular level, tacrolimus-induced impairment of mitochondrial respiration was significantly improved by coenzyme Q10, as evidenced by the increased mitochondrial oxygen consumption and ATP production. Our data indicate that coenzyme Q10 plays an important role in reducing tacrolimus-induced oxidative stress and protects the mitochondria in pancreatic beta cells. These findings suggest that supplementation with coenzyme Q10 has beneficial effects in tacrolimus-induced diabetes mellitus.
Collapse
Affiliation(s)
- Kang Luo
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Yu
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| | - Yi Quan
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Yoo Jin Shin
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Hong Lim Kim
- Integrative Research Support Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Eun Jeong Ko
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| | - Sun Woo Lim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea. .,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Park C, Kwon DH, Hwang SJ, Han MH, Jeong JW, Hong SH, Cha HJ, Hong SH, Kim GY, Lee HJ, Kim S, Kim HS, Choi YH. Protective Effects of Nargenicin A1 against Tacrolimus-Induced Oxidative Stress in Hirame Natural Embryo Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16061044. [PMID: 30909475 PMCID: PMC6466173 DOI: 10.3390/ijerph16061044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 12/13/2022]
Abstract
Tacrolimus is widely used as an immunosuppressant to reduce the risk of rejection after organ transplantation, but its cytotoxicity is problematic. Nargenicin A1 is an antibiotic extracted from Nocardia argentinensis and is known to have antioxidant activity, though its mode of action is unknown. The present study was undertaken to evaluate the protective effects of nargenicin A1 on DNA damage and apoptosis induced by tacrolimus in hirame natural embryo (HINAE) cells. We found that reduced HINAE cell survival by tacrolimus was due to the induction of DNA damage and apoptosis, both of which were prevented by co-treating nargenicin A1 or N-acetyl-l-cysteine, a reactive oxygen species (ROS) scavenger, with tacrolimus. In addition, apoptosis induction by tacrolimus was accompanied by increases in ROS generation and decreases in adenosine triphosphate (ATP) levels caused by mitochondrial dysfunction, and these changes were significantly attenuated in the presence of nargenicin A1, which further indicated tacrolimus-induced apoptosis involved an oxidative stress-associated mechanism. Furthermore, nargenicin A1 suppressed tacrolimus-induced B-cell lymphoma-2 (Bcl-2) down-regulation, Bax up-regulation, and caspase-3 activation. Collectively, these results demonstrate that nargenicin A1 protects HINAE cells against tacrolimus-induced DNA damage and apoptosis, at least in part, by scavenging ROS and thus suppressing the mitochondrial-dependent apoptotic pathway.
Collapse
Affiliation(s)
- Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dong-eui University, Busan 47340, Korea.
| | - Da Hye Kwon
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea.
| | - Su Jung Hwang
- Department of Pharmacy, College of Pharmacy, Inje University, Gimhae 50834, Korea.
| | - Min Ho Han
- National Marine Biodiversity Institute of Korea, Seocheon 33662, Korea.
| | - Jin-Woo Jeong
- Nakdonggang National Institute of Biological Resources, Sangju 17104, Korea.
| | - Sang Hoon Hong
- Department of Internal Medicine, Dong-eui University College of Korean Medicine, Busan 47227, Korea.
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea.
| | - Su-Hyun Hong
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea.
| | - Gi-Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea.
| | - Hyo-Jong Lee
- Department of Pharmacy, College of Pharmacy, Inje University, Gimhae 50834, Korea.
| | - Suhkmann Kim
- Department of Chemistry, College of Natural Sciences, Center for Proteome Biophysics and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Korea.
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Korea.
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea.
| |
Collapse
|
23
|
Saccà SC, Corazza P, Gandolfi S, Ferrari D, Sukkar S, Iorio EL, Traverso CE. Substances of Interest That Support Glaucoma Therapy. Nutrients 2019; 11:E239. [PMID: 30678262 PMCID: PMC6412416 DOI: 10.3390/nu11020239] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 12/14/2022] Open
Abstract
Glaucoma is a multifactorial disease in which pro-apoptotic signals are directed to retinal ganglion cells. During this disease the conventional outflow pathway becomes malfunctioning. Aqueous humour builds up in the anterior chamber, leading to increased intraocular pressure. Both of these events are related to functional impairment. The knowledge of molecular mechanisms allows us to better understand the usefulness of substances that can support anti-glaucoma therapy. The goal of glaucoma therapy is not simply to lower intraocular pressure; it should also be to facilitate the survival of retinal ganglion cells, as these constitute the real target tissue in this disease, in which the visual pathway is progressively compromised. Indeed, an endothelial dysfunction syndrome affecting the endothelial cells of the trabecular meshwork occurs in both normal-tension glaucoma and high-tension glaucoma. Some substances, such as polyunsaturated fatty acids, can counteract the damage due to the molecular mechanisms - whether ischemic, oxidative, inflammatory or other - that underlie the pathogenesis of glaucoma. In this review, we consider some molecules, such as polyphenols, that can contribute, not only theoretically, to neuroprotection but which are also able to counteract the metabolic pathways that lead to glaucomatous damage. Ginkgo biloba extract, for instance, improves the blood supply to peripheral districts, including the optic nerve and retina and exerts a neuro-protective action by inhibiting apoptosis. Polyunsaturated fatty acids can protect the endothelium and polyphenols exert an anti-inflammatory action through the down-regulation of cytokines such as TNF-α and IL-6. All these substances can aid anti-glaucoma therapy by providing metabolic support for the cells involved in glaucomatous injury. Indeed, it is known that the food we eat is able to change our gene expression.
Collapse
Affiliation(s)
- Sergio Claudio Saccà
- Ophthalmology Unit, Department of Head/Neck Pathologies, Policlinico San Martino Hospital, IRCCS Hospital-University San Martino, Viale Benedetto XV, 16132 Genoa, Italy.
| | - Paolo Corazza
- Eye Clinic, Department of Neuroscience and Sensory Organs, University of Genoa, Policlinico San Martino Hospital IRCCS Hospital-University San Martino, Viale Benedetto XV, 16132 Genoa, Italy.
| | - Stefano Gandolfi
- Ophthalmology Unit, Department of Biological, Biotechnological and Translational Sciences, University of Parma, 43121 Parma, Italy.
| | - Daniele Ferrari
- Ophthalmology Unit, Department of Head/Neck Pathologies, Policlinico San Martino Hospital, IRCCS Hospital-University San Martino, Viale Benedetto XV, 16132 Genoa, Italy.
| | - Samir Sukkar
- U.O. di Dietetica e Nutrizione Clinica, Policlinico San Martino Hospital IRCCS Hospital-University San Martino, 35122 Genoa, Italy.
| | - Eugenio Luigi Iorio
- International Observatory of Oxidative Stress, Via Paolo Grisignano 21, 84127 Salerno, Italy.
| | - Carlo Enrico Traverso
- Eye Clinic, Department of Neuroscience and Sensory Organs, University of Genoa, Policlinico San Martino Hospital IRCCS Hospital-University San Martino, Viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|
24
|
Abstract
BACKGROUND The effect of conversion to cytotoxic T lymphocyte-associated protein 4 immunoglobulin (CTLA4Ig) treatment on tacrolimus (TAC)-induced renal dysfunction is well known, but its effect on TAC-induced diabetes mellitus (DM) is still undetermined. In the present study, we tested the diabetogenicity of CTLA4Ig and evaluated the effect of conversion to CTLA4Ig treatment on TAC-induced diabetic rats. METHODS We tested diabetogenicity of CTLA4Ig by escalating doses (0.25, 0.5, 1, 2, and 4 mg/kg weekly) for 4 weeks. In the conversion study, we administered TAC (1.5 mg/kg) for 3 weeks and confirmed TAC-induced DM by intraperitoneal glucose tolerance test. Thereafter, TAC administration was continued, withdrawn, or replaced by CTLA4Ig treatment (1 or 2 mg/kg) for additional 3 weeks. The effect of CTLA4Ig on TAC-induced DM in vivo and in vitro was evaluated by assessing pancreatic islet function, histopathology, oxidative stress, apoptosis, and macrophage infiltration. RESULTS Intraperitoneal glucose tolerance test in the CTLA4Ig groups did not differ from the control group. In addition, plasma insulin level, glucose-induced insulin secretion, and islet viability were not different between the CTLA4Ig and control groups. In the conversion study, TAC withdrawal ameliorated pancreatic islet dysfunction compared with the TAC group, and conversion to CTLA4Ig further improved pancreatic islet function compared with the TAC withdrawal group. TAC-induced oxidative stress, apoptotic cell death, and infiltration of macrophages decreased with TAC withdrawal, and CTLA4Ig conversion further reduced those values. In the in vitro study, CTLA4Ig decreased TAC-induced pancreatic islet cell death and reactive oxygen species production. CONCLUSIONS CTLA4Ig was not diabetogenic, and conversion to CTLA4Ig reduced TAC-induced pancreatic islet injury.
Collapse
|
25
|
Li L, Qi J, Li H. Natural Products Modulating Autophagy Pathway Against the Pathogenesis of Diabetes Mellitus. Curr Drug Targets 2018; 20:96-110. [DOI: 10.2174/1389450119666180726115805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/07/2018] [Accepted: 06/21/2018] [Indexed: 01/19/2023]
Abstract
Autophagy is a conserved, regulated cellular process for the degradation of abnormal proteins
and disrupted organelles. Literature has described that dysregulation of autophagy is closely related
to the pathogenesis of diabetes mellitus in processes such as impaired pancreatic β cells function,
peripheral insulin resistance and diabetic complications. Emerging evidence indicates that natural
products may possess anti-diabetic activity via regulation of autophagy. In this review, we summarize
natural products targeting the pathogenesis of diabetes mellitus through the regulation of autophagy
and underline possible mechanisms, providing potential drug candidates or therapies for the treatment
of diabetes mellitus.
Collapse
Affiliation(s)
- Linghuan Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jiameng Qi
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hanbing Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
26
|
Lim SW, Shin YJ, Luo K, Quan Y, Ko EJ, Chung BH, Yang CW. Effect of Klotho on autophagy clearance in tacrolimus‐induced renal injury. FASEB J 2018; 33:2694-2706. [DOI: 10.1096/fj.201800751r] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Sun Woo Lim
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
| | - Yoo Jin Shin
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
| | - Kang Luo
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
| | - Yi Quan
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
| | - Eun Jeong Ko
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
- Division of NephrologyDepartment of Internal MedicineSeoul St. Mary's HospitalCollege of MedicineCatholic University of KoreaSeoulSouth Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
- Division of NephrologyDepartment of Internal MedicineSeoul St. Mary's HospitalCollege of MedicineCatholic University of KoreaSeoulSouth Korea
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic DiseaseSeoul St. Mary's HospitalCatholic University of KoreaSeoulSouth Korea
- Transplant Research CenterCatholic University of KoreaSeoulSouth Korea
- Division of NephrologyDepartment of Internal MedicineSeoul St. Mary's HospitalCollege of MedicineCatholic University of KoreaSeoulSouth Korea
| |
Collapse
|