51
|
Jalgaonkar MP, Parmar UM, Kulkarni YA, Oza MJ. SIRT1-FOXOs activity regulates diabetic complications. Pharmacol Res 2021; 175:106014. [PMID: 34856334 DOI: 10.1016/j.phrs.2021.106014] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023]
Abstract
The prevalence of diabetes is continuously increasing in the recent decades. Persistent hyperglycemia, hyperinsulinemia and the subsequent oxidative stress result in diabetic complications, primarily categorized as microvascular (nephropathy, retinopathy and neuropathy) and macrovascular (cardiomyopathy) complications. The complications are prevalent in both type 1 and type 2 diabetic patients. Polyol pathway, elevated AGE production, PKC activation and hexosamine pathway are indeed the critical pathways involved in the progression of diabetic complications. Silent information regulator 2 or SIR2 or more commonly known as sirtuins are NAD+ dependent histone deacetylase. SIRT1, a member of the sirtuin family has been extensively studied for its role in lifespan extension and needs to be explored for its beneficial effects in diabetic complications. Moreover, it is also known to regulate the activity of other proteins and transcription factors. One such substrate of SIRT1 is FOXOs transcription factor which has gained much attention as the mediator of various cellular processes such as cell cycle arrest and proliferation, DNA repair and metabolism. It has been reported that SIRT1 regulates the activity of FOXOs, whereas few recent advances also suggest a role FOXOs in governing the activity of SIRT1, which permits for a crosstalk between SIRT1 and FOXOs. Therefore, the focus of the present review is to describe and explore the interaction between SIRT1 and FOXOs, predominantly FOXO1 and FOXO3 and to understand the underlying mechanism of SIRT1-FOXOs in controlling and alleviating diabetic complications. Thus, this crosstalk suggests that SIRT1 and FOXOs may serve as potential therapeutic targets in treating diabetic complications.
Collapse
Affiliation(s)
- Manjiri P Jalgaonkar
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India
| | - Urvi M Parmar
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai 400056, India
| | - Manisha J Oza
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|
52
|
Upadhyay A. Natural compounds in the regulation of proteostatic pathways: An invincible artillery against stress, ageing, and diseases. Acta Pharm Sin B 2021; 11:2995-3014. [PMID: 34729300 PMCID: PMC8546668 DOI: 10.1016/j.apsb.2021.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/12/2020] [Accepted: 11/03/2020] [Indexed: 01/13/2023] Open
Abstract
Cells have different sets of molecules for performing an array of physiological functions. Nucleic acids have stored and carried the information throughout evolution, whereas proteins have been attributed to performing most of the cellular functions. To perform these functions, proteins need to have a unique conformation and a definite lifespan. These attributes are achieved by a highly coordinated protein quality control (PQC) system comprising chaperones to fold the proteins in a proper three-dimensional structure, ubiquitin-proteasome system for selective degradation of proteins, and autophagy for bulk clearance of cell debris. Many kinds of stresses and perturbations may lead to the weakening of these protective cellular machinery, leading to the unfolding and aggregation of cellular proteins and the occurrence of numerous pathological conditions. However, modulating the expression and functional efficiency of molecular chaperones, E3 ubiquitin ligases, and autophagic proteins may diminish cellular proteotoxic load and mitigate various pathological effects. Natural medicine and small molecule-based therapies have been well-documented for their effectiveness in modulating these pathways and reestablishing the lost proteostasis inside the cells to combat disease conditions. The present article summarizes various similar reports and highlights the importance of the molecules obtained from natural sources in disease therapeutics.
Collapse
Key Words
- 17-AAG, 17-allylamino-geldanamycin
- APC, anaphase-promoting complex
- Ageing
- Autophagy
- BAG, BCL2-associated athanogene
- CAP, chaperone-assisted proteasomal degradation
- CASA, chaperone-assisted selective autophagy
- CHIP, carboxy-terminus of HSC70 interacting protein
- CMA, chaperone-mediated autophagy
- Cancer
- Chaperones
- DUBs, deubiquitinases
- Drug discovery
- EGCG, epigallocatechin-3-gallate
- ESCRT, endosomal sorting complexes required for transport
- HECT, homologous to the E6-AP carboxyl terminus
- HSC70, heat shock cognate 70
- HSF1, heat shock factor 1
- HSP, heat shock protein
- KFERQ, lysine-phenylalanine-glutamate-arginine-glutamine
- LAMP2a, lysosome-associated membrane protein 2a
- LC3, light chain 3
- NBR1, next to BRCA1 gene 1
- Natural molecules
- Neurodegeneration
- PQC, protein quality control
- Proteinopathies
- Proteostasis
- RING, really interesting new gene
- UPS, ubiquitin–proteasome system
- Ub, ubiquitin
- Ubiquitin proteasome system
Collapse
Affiliation(s)
- Arun Upadhyay
- Department of Biochemistry, Central University of Rajasthan, Bandar Sindari, Kishangarh, Ajmer, Rajasthan 305817, India
| |
Collapse
|
53
|
Brimson JM, Prasanth MI, Malar DS, Thitilertdecha P, Kabra A, Tencomnao T, Prasansuklab A. Plant Polyphenols for Aging Health: Implication from Their Autophagy Modulating Properties in Age-Associated Diseases. Pharmaceuticals (Basel) 2021; 14:ph14100982. [PMID: 34681206 PMCID: PMC8538309 DOI: 10.3390/ph14100982] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/13/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
Polyphenols are a family of naturally occurring organic compounds, majorly present in fruits, vegetables, and cereals, characterised by multiple phenol units, including flavonoids, tannic acid, and ellagitannin. Some well-known polyphenols include resveratrol, quercetin, curcumin, epigallocatechin gallate, catechin, hesperetin, cyanidin, procyanidin, caffeic acid, and genistein. They can modulate different pathways inside the host, thereby inducing various health benefits. Autophagy is a conserved process that maintains cellular homeostasis by clearing the damaged cellular components and balancing cellular survival and overall health. Polyphenols could maintain autophagic equilibrium, thereby providing various health benefits in mediating neuroprotection and exhibiting anticancer and antidiabetic properties. They could limit brain damage by dismantling misfolded proteins and dysfunctional mitochondria, thereby activating autophagy and eliciting neuroprotection. An anticarcinogenic mechanism is stimulated by modulating canonical and non-canonical signalling pathways. Polyphenols could also decrease insulin resistance and inhibit loss of pancreatic islet β-cell mass and function from inducing antidiabetic activity. Polyphenols are usually included in the diet and may not cause significant side effects that could be effectively used to prevent and treat major diseases and ailments.
Collapse
Affiliation(s)
- James Michael Brimson
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10330, Thailand;
| | - Atul Kabra
- Department of Pharmacology, University Institute of Pharma Sciences, Chandigarh University, Sahibzad Ajit Singh Nagar 140413, Punjab, India;
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (T.T.); (A.P.)
| | - Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- College of Public Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (T.T.); (A.P.)
| |
Collapse
|
54
|
Sun X, Wang L, Obayomi SMB, Wei Z. Epigenetic Regulation of β Cell Identity and Dysfunction. Front Endocrinol (Lausanne) 2021; 12:725131. [PMID: 34630329 PMCID: PMC8498190 DOI: 10.3389/fendo.2021.725131] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/08/2021] [Indexed: 01/07/2023] Open
Abstract
β cell dysfunction and failure are driving forces of type 2 diabetes mellitus (T2DM) pathogenesis. Investigating the underlying mechanisms of β cell dysfunction may provide novel targets for the development of next generation therapy for T2DM. Epigenetics is the study of gene expression changes that do not involve DNA sequence changes, including DNA methylation, histone modification, and non-coding RNAs. Specific epigenetic signatures at all levels, including DNA methylation, chromatin accessibility, histone modification, and non-coding RNA, define β cell identity during embryonic development, postnatal maturation, and maintain β cell function at homeostatic states. During progression of T2DM, overnutrition, inflammation, and other types of stress collaboratively disrupt the homeostatic epigenetic signatures in β cells. Dysregulated epigenetic signatures, and the associating transcriptional outputs, lead to the dysfunction and eventual loss of β cells. In this review, we will summarize recent discoveries of the establishment and disruption of β cell-specific epigenetic signatures, and discuss the potential implication in therapeutic development.
Collapse
Affiliation(s)
- Xiaoqiang Sun
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, United States
- Tianjin Fourth Central Hospital, Tianjin, China
- The Fourth Central Hospital Affiliated to Nankai University, Tianjin, China
- The Fourth Central Hospital Clinical College, Tianjin Medical University, Tianjin, China
| | - Liu Wang
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - S M Bukola Obayomi
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Zong Wei
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, United States
| |
Collapse
|
55
|
Bharath LP, Rockhold JD, Conway R. Selective Autophagy in Hyperglycemia-Induced Microvascular and Macrovascular Diseases. Cells 2021; 10:cells10082114. [PMID: 34440882 PMCID: PMC8392047 DOI: 10.3390/cells10082114] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/07/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of autophagy is an important underlying cause in the onset and progression of many metabolic diseases, including diabetes. Studies in animal models and humans show that impairment in the removal and the recycling of organelles, in particular, contributes to cellular damage, functional failure, and the onset of metabolic diseases. Interestingly, in certain contexts, inhibition of autophagy can be protective. While the inability to upregulate autophagy can play a critical role in the development of diseases, excessive autophagy can also be detrimental, making autophagy an intricately regulated process, the altering of which can adversely affect organismal health. Autophagy is indispensable for maintaining normal cardiac and vascular structure and function. Patients with diabetes are at a higher risk of developing and dying from vascular complications. Autophagy dysregulation is associated with the development of heart failure, many forms of cardiomyopathy, atherosclerosis, myocardial infarction, and microvascular complications in diabetic patients. Here, we review the recent findings on selective autophagy in hyperglycemia and diabetes-associated microvascular and macrovascular complications.
Collapse
|
56
|
Chen X, Le Y, He WY, He J, Wang YH, Zhang L, Xiong QM, Zheng XQ, Liu KX, Wang HB. Abnormal Insulin-like Growth Factor 1 Signaling Regulates Neuropathic Pain by Mediating the Mechanistic Target of Rapamycin-Related Autophagy and Neuroinflammation in Mice. ACS Chem Neurosci 2021; 12:2917-2928. [PMID: 34264648 DOI: 10.1021/acschemneuro.1c00271] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neuropathic pain is a chronic condition with little specific treatment. Insulin-like growth factor 1 (IGF1), interacting with its receptor, IGF1R, serves a vital role in neuronal and brain functions such as autophagy and neuroinflammation. Yet, the function of spinal IGF1/IGF1R in neuropathic pain is unclear. Here, we examined whether and how spinal IGF1 signaling affects pain-like behaviors in mice with chronic constriction injury (CCI) of the sciatic nerve. To corroborate the role of IGF1, we injected intrathecally IGF1R inhibitor (nvp-aew541) or anti-IGF1 neutralizing antibodies. We found that IGF1 (derived from astrocytes) in the lumbar cord increased along with the neuropathic pain induced by CCI. IGF1R was predominantly expressed on neurons. IGF1R antagonism or IGF1 neutralization attenuated pain behaviors induced by CCI, relieved mTOR-related suppression of autophagy, and mitigated neuroinflammation in the spinal cord. These findings reveal that the abnormal IGF1/IGF1R signaling contributes to neuropathic pain by exacerbating autophagy dysfunction and neuroinflammation.
Collapse
Affiliation(s)
- Xin Chen
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Yue Le
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Wan-you He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Jian He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Yun-hua Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Lei Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Qing-ming Xiong
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Xue-qin Zheng
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Ke-xuan Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Han-bing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| |
Collapse
|
57
|
Sun-Wang JL, Yarritu-Gallego A, Ivanova S, Zorzano A. The ubiquitin-proteasome system and autophagy: self-digestion for metabolic health. Trends Endocrinol Metab 2021; 32:594-608. [PMID: 34034951 DOI: 10.1016/j.tem.2021.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 01/02/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a global health challenge. Therefore, understanding the molecular mechanisms underlying the pathophysiology of T2DM is key to improving current therapies. Loss of protein homeostasis leads to the accumulation of damaged proteins in cells, which results in tissue dysfunction. The elimination of damaged proteins occurs through the ubiquitin-proteasome system (UPS) and autophagy. In this review, we describe the mutual regulation between the UPS and autophagy and the involvement of these two proteolytic systems in metabolic dysregulation, insulin resistance, and T2DM. We propose that alterations in the UPS or autophagy contribute to triggering insulin resistance and the development of T2DM. In addition, these two pathways emerge as promising therapeutic targets for improving insulin resistance.
Collapse
Affiliation(s)
- Jia Liang Sun-Wang
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.
| | - Alex Yarritu-Gallego
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany; Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Saška Ivanova
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain.
| |
Collapse
|
58
|
Dewanjee S, Vallamkondu J, Kalra RS, John A, Reddy PH, Kandimalla R. Autophagy in the diabetic heart: A potential pharmacotherapeutic target in diabetic cardiomyopathy. Ageing Res Rev 2021; 68:101338. [PMID: 33838320 DOI: 10.1016/j.arr.2021.101338] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022]
Abstract
Association of diabetes with an elevated risk of cardiac failure has been clinically evident. Diabetes potentiates diastolic and systolic cardiac failure following the myocardial infarction that produces the cardiac muscle-specific microvascular complication, clinically termed as diabetic cardiomyopathy. Elevated susceptibility of diabetic cardiomyopathy is primarily caused by the generation of free radicals in the hyperglycemic milieu, compromising the myocardial contractility and normal cardiac functions with increasing redox insult, impaired mitochondria, damaged organelles, apoptosis, and cardiomyocytes fibrosis. Autophagy is essentially involved in the recycling/clearing the damaged organelles, cytoplasmic contents, and aggregates, which are frequently produced in cardiomyocytes. Although autophagy plays a vital role in maintaining the cellular homeostasis in diligent cardiac tissues, this process is frequently impaired in the diabetic heart. Given its clinical significance, accumulating evidence largely showed the functional aspects of autophagy in diabetic cardiomyopathy, elucidating its intricate protective and pathogenic outcomes. However, etiology and molecular readouts of these contrary autophagy activities in diabetic cardiomyopathy are not yet comprehensively assessed and translated. In this review, we attempted to assess the role of autophagy and its adaptations in the diabetic heart. To delineate the molecular consequences of these events, we provided detailed insights into the autophagy regulation pieces of machinery including the mTOR/AMPK, TFEB/ZNSCAN3, FOXOs, SIRTs, PINK1/Parkin, Nrf2, miRNAs, and others in the diabetic cardiomyopathy. Given the clinical significance of autophagy in the diabetic heart, we further discussed the potential pharmacotherapeutic strategies towards targeting autophagy. Taken together, the present report meticulously assessed autophagy, its adaptations, and molecular regulations in diabetic cardiomyopathy and reviewed the current autophagy-targeting strategies.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | | | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba, 305 8565, Japan.
| | - Albin John
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, 506007, Telangana, India; Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad, 50000, Telangana, India.
| |
Collapse
|
59
|
Li X, Wan T, Li Y. Role of FoxO1 in regulating autophagy in type 2 diabetes mellitus (Review). Exp Ther Med 2021; 22:707. [PMID: 34007316 PMCID: PMC8120662 DOI: 10.3892/etm.2021.10139] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major chronic disease that is characterized by pancreatic β-cell dysfunction and insulin resistance. Autophagy is a highly conserved intracellular recycling pathway and is involved in regulating intracellular homeostasis. Transcription factor Forkhead box O1 (FoxO1) also regulates fundamental cellular processes, including cell differentiation, metabolism and apoptosis, and proliferation to cellular stress. Increasing evidence suggest that autophagy and FoxO1 are involved in the pathogenesis of T2DM, including β-cell viability, apoptosis, insulin secretion and peripheral insulin resistance. Recent studies have demonstrated that FoxO1 improves insulin resistance by regulating target tissue autophagy. The present review summarizes current literature on the role of autophagy and FoxO1 in T2DM. The participation of FoxO1 in the development and occurrence of T2DM via autophagy is also discussed.
Collapse
Affiliation(s)
- Xiudan Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tingting Wan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yanbo Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
60
|
Byrne NJ, Rajasekaran NS, Abel ED, Bugger H. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radic Biol Med 2021; 169:317-342. [PMID: 33910093 PMCID: PMC8285002 DOI: 10.1016/j.freeradbiomed.2021.03.046] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Even in the absence of coronary artery disease and hypertension, diabetes mellitus (DM) may increase the risk for heart failure development. This risk evolves from functional and structural alterations induced by diabetes in the heart, a cardiac entity termed diabetic cardiomyopathy (DbCM). Oxidative stress, defined as the imbalance of reactive oxygen species (ROS) has been increasingly proposed to contribute to the development of DbCM. There are several sources of ROS production including the mitochondria, NAD(P)H oxidase, xanthine oxidase, and uncoupled nitric oxide synthase. Overproduction of ROS in DbCM is thought to be counterbalanced by elevated antioxidant defense enzymes such as catalase and superoxide dismutase. Excess ROS in the cardiomyocyte results in further ROS production, mitochondrial DNA damage, lipid peroxidation, post-translational modifications of proteins and ultimately cell death and cardiac dysfunction. Furthermore, ROS modulates transcription factors responsible for expression of antioxidant enzymes. Lastly, evidence exists that several pharmacological agents may convey cardiovascular benefit by antioxidant mechanisms. As such, increasing our understanding of the pathways that lead to increased ROS production and impaired antioxidant defense may enable the development of therapeutic strategies against the progression of DbCM. Herein, we review the current knowledge about causes and consequences of ROS in DbCM, as well as the therapeutic potential and strategies of targeting oxidative stress in the diabetic heart.
Collapse
Affiliation(s)
- Nikole J Byrne
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
61
|
Barteková M, Adameová A, Görbe A, Ferenczyová K, Pecháňová O, Lazou A, Dhalla NS, Ferdinandy P, Giricz Z. Natural and synthetic antioxidants targeting cardiac oxidative stress and redox signaling in cardiometabolic diseases. Free Radic Biol Med 2021; 169:446-477. [PMID: 33905865 DOI: 10.1016/j.freeradbiomed.2021.03.045] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiometabolic diseases (CMDs) are metabolic diseases (e.g., obesity, diabetes, atherosclerosis, rare genetic metabolic diseases, etc.) associated with cardiac pathologies. Pathophysiology of most CMDs involves increased production of reactive oxygen species and impaired antioxidant defense systems, resulting in cardiac oxidative stress (OxS). To alleviate OxS, various antioxidants have been investigated in several diseases with conflicting results. Here we review the effect of CMDs on cardiac redox homeostasis, the role of OxS in cardiac pathologies, as well as experimental and clinical data on the therapeutic potential of natural antioxidants (including resveratrol, quercetin, curcumin, vitamins A, C, and E, coenzyme Q10, etc.), synthetic antioxidants (including N-acetylcysteine, SOD mimetics, mitoTEMPO, SkQ1, etc.), and promoters of antioxidant enzymes in CMDs. As no antioxidant indicated for the prevention and/or treatment of CMDs has reached the market despite the large number of preclinical and clinical studies, a sizeable translational gap is evident in this field. Thus, we also highlight potential underlying factors that may contribute to the failure of translation of antioxidant therapies in CMDs.
Collapse
Affiliation(s)
- Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia.
| | - Adriana Adameová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Kristína Ferenczyová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Oľga Pecháňová
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, 81371 Bratislava, Slovakia
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, And Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
62
|
Dewanjee S, Vallamkondu J, Kalra RS, Chakraborty P, Gangopadhyay M, Sahu R, Medala V, John A, Reddy PH, De Feo V, Kandimalla R. The Emerging Role of HDACs: Pathology and Therapeutic Targets in Diabetes Mellitus. Cells 2021; 10:1340. [PMID: 34071497 PMCID: PMC8228721 DOI: 10.3390/cells10061340] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus (DM) is one of the principal manifestations of metabolic syndrome and its prevalence with modern lifestyle is increasing incessantly. Chronic hyperglycemia can induce several vascular complications that were referred to be the major cause of morbidity and mortality in DM. Although several therapeutic targets have been identified and accessed clinically, the imminent risk of DM and its prevalence are still ascending. Substantial pieces of evidence revealed that histone deacetylase (HDAC) isoforms can regulate various molecular activities in DM via epigenetic and post-translational regulation of several transcription factors. To date, 18 HDAC isoforms have been identified in mammals that were categorized into four different classes. Classes I, II, and IV are regarded as classical HDACs, which operate through a Zn-based mechanism. In contrast, class III HDACs or Sirtuins depend on nicotinamide adenine dinucleotide (NAD+) for their molecular activity. Functionally, most of the HDAC isoforms can regulate β cell fate, insulin release, insulin expression and signaling, and glucose metabolism. Moreover, the roles of HDAC members have been implicated in the regulation of oxidative stress, inflammation, apoptosis, fibrosis, and other pathological events, which substantially contribute to diabetes-related vascular dysfunctions. Therefore, HDACs could serve as the potential therapeutic target in DM towards developing novel intervention strategies. This review sheds light on the emerging role of HDACs/isoforms in diabetic pathophysiology and emphasized the scope of their targeting in DM for constituting novel interventional strategies for metabolic disorders/complications.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | | | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba 305 8565, Japan;
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | - Moumita Gangopadhyay
- School of Life Science and Biotechnology, ADAMAS University, Barasat, Kolkata 700126, West Bengal, India;
| | - Ranabir Sahu
- Department of Pharmaceutical Technology, University of North Bengal, Darjeeling 734013, West Bengal, India;
| | - Vijaykrishna Medala
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India;
| | - Albin John
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.J.); (P.H.R.)
| | - P. Hemachandra Reddy
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.J.); (P.H.R.)
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India;
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana, India
| |
Collapse
|
63
|
Qindan Capsule Attenuates Myocardial Hypertrophy and Fibrosis in Pressure Overload-Induced Mice Involving mTOR and TGF- β1/Smad Signaling Pathway Inhibition. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5577875. [PMID: 34007292 PMCID: PMC8102107 DOI: 10.1155/2021/5577875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/03/2021] [Accepted: 04/20/2021] [Indexed: 01/23/2023]
Abstract
Qindan capsule (QC), a traditional Chinese medicine compound, has been used to treat hypertension in the clinic for over 30 years. It is still not known about the effects of QC on pressure overload-induced cardiac remodeling. Hence, this study aims to investigate the effects of QC on pressure overload-induced cardiac hypertrophy, fibrosis, and heart failure in mice and to determine the possible mechanisms. Transverse aortic constriction (TAC) surgery was used to induce cardiac hypertrophy and heart failure in C57BL/6 mice. Mice were treated with QC or losartan for 8 weeks after TAC surgery. Cardiac function indexes were evaluated with transthoracic echocardiography. Cardiac pathology was detected using HE and Masson's trichrome staining. Cardiomyocyte ultrastructure was detected using transmission electron microscopy. Hypertrophy-related fetal gene expression was investigated using real-time RT-PCR. The expression of 8-OHdG and the concentration of MDA and Ang-II were assessed by immunohistochemistry stain and ELISA assay, respectively. The total and phosphorylated protein levels of mTOR, p70S6K, 4EBP1, Smad2, and Smad3 and the expression of TGF-β1 and collagen I were measured using western blot. The results showed that low- and high-dose QC improved pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction. QC inhibited ANP, BNP, and β-MHC mRNA expression in failing hearts. QC improved myocardial ultrastructure after TAC surgery. Furthermore, QC downregulated the expression of 8-OHdG and the concentration of MDA, 15-F2t-IsoP, and Ang-II in heart tissues after TAC surgery. We also found that QC inhibited the phosphorylation of mTOR, p70S6K, and 4EBP1 and the expression of TGF-β1, p-Smad2, p-Smad3, and collagen I in pressure overload-induced failing hearts. These data indicate that QC has direct benefic effects on pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction. The protective effects of QC involve prevention of increased oxidative stress injury and Ang-II levels and inhibition of mTOR and TGF-β1/Smad pathways in failing hearts.
Collapse
|
64
|
PXDN reduces autophagic flux in insulin-resistant cardiomyocytes via modulating FoxO1. Cell Death Dis 2021; 12:418. [PMID: 33903591 PMCID: PMC8076187 DOI: 10.1038/s41419-021-03699-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022]
Abstract
Autophagy, a well-observed intracellular lysosomal degradation process, is particularly important to the cell viability in diabetic cardiomyopathy (DCM). Peroxidasin (PXDN) is a heme-containing peroxidase that augments oxidative stress and plays an essential role in cardiovascular diseases, while whether PXDN contributes to the pathogenesis of DCM remains unknown. Here we reported the suppression of cell viability and autophagic flux, as shown by autophagosomes accumulation and increased expression level of LC3-II and p62 in cultured H9C2 and human AC16 cells that treated with 400 μM palmitate acid (PA) for 24 h. Simultaneously, PXDN protein level increased. Moreover, cell death, autophagosomes accumulation as well as increased p62 expression were suppressed by PXDN silence. In addition, knockdown of PXDN reversed PA-induced downregulated forkhead box-1 (FoxO1) and reduced FoxO1 phosphorylation, whereas did not affect AKT phosphorylation. Not consistent with the effects of si-PXDN, double-silence of PXDN and FoxO1 significantly increased cell death, suppressed autophagic flux and declined the level of FoxO1 and PXDN, while the expression of LC3-II was unchanged under PA stimulation. Furthermore, inhibition of FoxO1 in PA-untreated cells induced cell death, inhibited autophagic flux, and inhibited FoxO1 and PXDN expression. Thus, we come to conclusion that PXDN plays a key role in PA-induced cell death by impairing autophagic flux through inhibiting FoxO1, and FoxO1 may also affect the expression of PXDN. These findings may develop better understanding of potential mechanisms regarding autophagy in insulin-resistant cardiomyocytes.
Collapse
|
65
|
Yang X, Song X, Li Z, Liu N, Yan Y, Liu B. Crosstalk between extracellular vesicles and autophagy in cardiovascular pathophysiology. Pharmacol Res 2021; 172:105628. [PMID: 33887437 DOI: 10.1016/j.phrs.2021.105628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/16/2021] [Indexed: 01/13/2023]
Abstract
Extracellular vesicles are composed of loaded soluble substances and lipid bilayers; these include apoptotic bodies, exosomes, and microvesicles. Extracellular vesicles, as carriers of biological information between cells, have been recognized for their role in the diagnosis and treatment of cardiovascular diseases. The biogenesis of extracellular vesicles is closely related to autophagy. Moreover, extracellular vesicles further affect autophagy levels in target cells through their transmitted contents. Autophagy is a catabolic cell process that maintains cell homeostasis by eliminating misfolded proteins and damaged organelles. Existing studies have revealed that extracellular vesicles and autophagy share molecular mechanisms with notable crosstalk, including, perspectives such as amphisomes and "secretory autophagy." In this review, we first introduce the biogenesis of extracellular vesicles and the classic views of autophagy before moving onto the crosstalk between extracellular vesicles and autophagy. Finally, we discuss the research progress of extracellular vesicles and autophagy in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Xingru Yang
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Xianjing Song
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Zhibo Li
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Ning Liu
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China.
| |
Collapse
|
66
|
Palomer X, Aguilar-Recarte D, García R, Nistal JF, Vázquez-Carrera M. Sirtuins: To Be or Not To Be in Diabetic Cardiomyopathy. Trends Mol Med 2021; 27:554-571. [PMID: 33839024 DOI: 10.1016/j.molmed.2021.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy is the leading cause of death among people with diabetes. Despite its severity and poor prognosis, there are currently no approved specific drugs to prevent or even treat diabetic cardiomyopathy. There is a need to understand the pathogenic mechanisms underlying the development of diabetic cardiomyopathy to design new therapeutic strategies. These mechanisms are complex and intricate and include metabolic dysregulation, inflammation, oxidative stress, fibrosis, and apoptosis. Sirtuins, a group of deacetylase enzymes, play an important role in all these processes and are, therefore, potential molecular targets for treating this disease. In this review, we discuss the role of sirtuins in the heart, focusing on their contribution to the pathogenesis of diabetic cardiomyopathy and how their modulation could be therapeutically useful.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - David Aguilar-Recarte
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Raquel García
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - J Francisco Nistal
- Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Departamento de Ciencias Médicas y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL); Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Santander, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
67
|
Sun J, Tai S, Tang L, Yang H, Chen M, Xiao Y, Li X, Zhu Z, Zhou S. Acetylation Modification During Autophagy and Vascular Aging. Front Physiol 2021; 12:598267. [PMID: 33828486 PMCID: PMC8019697 DOI: 10.3389/fphys.2021.598267] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular aging plays a pivotal role in the morbidity and mortality of elderly people. Decrease in autophagy leads to acceleration of vascular aging, while increase in autophagy leads to deceleration of vascular aging. And emerging evidence indicates that acetylation plays an important role in autophagy regulation; therefore, recent research has focused on an in-depth analysis of the mechanisms underlying this regulation. In this review, current knowledge on the role of acetylation of autophagy-related proteins and the mechanisms by which acetylation including non-autophagy-related acetylation and autophagy related acetylation regulate vascular aging have been discussed. We conclude that the occurrence of acetylation modification during autophagy is a fundamental mechanism underlying autophagy regulation and provides promising targets to retard vascular aging.
Collapse
Affiliation(s)
- Jiaxing Sun
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shi Tai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liang Tang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Yang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mingxian Chen
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yichao Xiao
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xuping Li
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhaowei Zhu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
68
|
Transcription factor EB (TFEB)-mediated autophagy protects bovine mammary epithelial cells against H 2O 2-induced oxidative damage in vitro. J Anim Sci Biotechnol 2021; 12:35. [PMID: 33685494 PMCID: PMC7941962 DOI: 10.1186/s40104-021-00561-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/25/2021] [Indexed: 01/01/2023] Open
Abstract
Background Bovine mammary epithelial cells after calving undergo serious metabolic challenges and oxidative stress both of which could compromise autophagy. Transcription factor EB (TFEB)-mediated autophagy is an important cytoprotective mechanism against oxidative stress. However, effects of TFEB-mediated autophagy on the oxidative stress of bovine mammary epithelial cells remain unknown. Therefore, the main aim of the study was to investigate the role of TFEB-mediated autophagy in bovine mammary epithelial cells experiencing oxidative stress. Results H2O2 challenge of the bovine mammary epithelial cell MAC-T increased protein abundance of LC3-II, increased number of autophagosomes and autolysosomes while decreased protein abundance of p62. Inhibition of autophagy via bafilomycin A1 aggravated H2O2-induced reactive oxygen species (ROS) accumulation and apoptosis in MAC-T cells. Furthermore, H2O2 treatment triggered the translocation of TFEB into the nucleus. Knockdown of TFEB by siRNA reversed the effect of H2O2 on protein abundance of LC3-II and p62 as well as the number of autophagosomes and autolysosomes. Overexpression of TFEB activated autophagy and attenuated H2O2-induced ROS accumulation. Furthermore, TFEB overexpression attenuated H2O2-induced apoptosis by downregulating the caspase apoptotic pathway. Conclusions Our results indicate that activation of TFEB mediated autophagy alleviates H2O2-induced oxidative damage by reducing ROS accumulation and inhibiting caspase-dependent apoptosis.
Collapse
|
69
|
Wu Y, Xun Y, Zhang J, Hu H, Qin B, Wang T, Wang S, Li C, Lu Y. Resveratrol Attenuates Oxalate-Induced Renal Oxidative Injury and Calcium Oxalate Crystal Deposition by Regulating TFEB-Induced Autophagy Pathway. Front Cell Dev Biol 2021; 9:638759. [PMID: 33718378 PMCID: PMC7947311 DOI: 10.3389/fcell.2021.638759] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
The oxidative injury of renal tubular epithelial cells caused by inflammation and oxidative stress induced by hyperoxaluria is an important factor in the kidney calcium oxalate (CaOx) stone formation. Resveratrol (RSV) has been reported to reduce oxidative injury to renal tubular epithelial cells, and autophagy is critical for the protective effect of resveratrol. However, the protective mechanism of RSV in oxalate-induced oxidative injury of renal tubular cells and the role of autophagy in this process are still unclear. In our study, glyoxylic acid monohydrate-induced rats were treated with or without resveratrol, and it was detected that the overexpression of oxidant species, CaOx crystal deposition, apoptosis level, inflammatory cytokines and osteoblastic-associated protein expression were reversed by resveratrol. Additionally, Resveratrol pretreatment significantly reversed oxalate -induced decline in cell viability, cell damage, oxidant species overexpression, and osteogenic transformation in normal rat kidney epithelial-like (NRK-52E) cells. Furthermore, we found that RSV pretreatment promoted intracellular LC3II upregulation, p62 downregulation, and autophagosome formation, whereas 3-methyladenine treatment reduced this effect. Moreover, RSV induced the expression of transcription factor EB (TFEB) in the nucleus of NRK-52E cells in a concentration-dependent manner. After transfection of NRK-52E cells with TFEB siRNA, we showed that the RSV-induced increase in TFEB expression and autophagosome formation were inhibited. Simultaneously, RSV-induced NRK-52E cells protection was partially reversed. These results suggested that RSV regulates oxalate-induced renal inflammation, oxidative injury, and CaOx crystal deposition in vitro and in vivo through the activation of a TFEB-induced autophagy.
Collapse
Affiliation(s)
- Yue Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Xun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Henglong Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baolong Qin
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchao Lu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
70
|
Dietary Polyphenols in Metabolic and Neurodegenerative Diseases: Molecular Targets in Autophagy and Biological Effects. Antioxidants (Basel) 2021; 10:antiox10020142. [PMID: 33498216 PMCID: PMC7908992 DOI: 10.3390/antiox10020142] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022] Open
Abstract
Polyphenols represent a group of secondary metabolites of plants which have been analyzed as potent regulators of multiple biological processes, including cell proliferation, apoptosis, and autophagy, among others. These natural compounds exhibit beneficial effects and protection against inflammation, oxidative stress, and related injuries including metabolic diseases, such as cardiovascular damage, obesity and diabetes, and neurodegeneration. This review aims to summarize the mechanisms of action of polyphenols in relation to the activation of autophagy, stimulation of mitochondrial function and antioxidant defenses, attenuation of oxidative stress, and reduction in cell apoptosis, which may be responsible of the health promoting properties of these compounds.
Collapse
|
71
|
Ke X, Lin Z, Ye Z, Leng M, Chen B, Jiang C, Jiang X, Li G. Histone Deacetylases in the Pathogenesis of Diabetic Cardiomyopathy. Front Endocrinol (Lausanne) 2021; 12:679655. [PMID: 34367065 PMCID: PMC8339406 DOI: 10.3389/fendo.2021.679655] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022] Open
Abstract
The global burden of diabetes mellitus and its complications are currently increasing. Diabetic cardiomyopathy (DCM) is the main cause of diabetes mellitus associated morbidity and mortality; therefore, a comprehensive understanding of DCM development is required for more effective treatment. A disorder of epigenetic posttranscriptional modification of histones in chromatin has been reported to be associated with the pathology of DCM. Recent studies have implicated that histone deacetylases could regulate cardiovascular and metabolic diseases in cellular processes including cardiac fibrosis, hypertrophy, oxidative stress and inflammation. Therefore in this review, we summarized the roles of histone deacetylases in the pathogenesis of DCM, aiming to provide insights into exploring potential preventative and therapeutic strategies of DCM.
Collapse
Affiliation(s)
- Xiangyu Ke
- Centre of Clinical Epidemiology and Methodology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhirui Lin
- Centre of Clinical Epidemiology and Methodology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zebing Ye
- Department of Cardiology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Meifang Leng
- Department of Cardiology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Bo Chen
- Department of Endocrinology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Chunjie Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyun Jiang
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Xiaoyun Jiang, ; Guowei Li,
| | - Guowei Li
- Centre of Clinical Epidemiology and Methodology, Guangdong Second Provincial General Hospital, Guangzhou, China
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- *Correspondence: Xiaoyun Jiang, ; Guowei Li,
| |
Collapse
|
72
|
Zang H, Wu W, Qi L, Tan W, Nagarkatti P, Nagarkatti M, Wang X, Cui T. Autophagy Inhibition Enables Nrf2 to Exaggerate the Progression of Diabetic Cardiomyopathy in Mice. Diabetes 2020; 69:2720-2734. [PMID: 32948607 PMCID: PMC7679777 DOI: 10.2337/db19-1176] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
Nuclear factor-erythroid factor 2-related factor 2 (Nrf2) may either ameliorate or worsen diabetic cardiomyopathy. However, the underlying mechanisms are poorly understood. Herein we report a novel mechanism of Nrf2-mediated myocardial damage in type 1 diabetes (T1D). Global Nrf2 knockout (Nrf2KO) hardly affected the onset of cardiac dysfunction induced by T1D but slowed down its progression in mice independent of sex. In addition, Nrf2KO inhibited cardiac pathological remodeling, apoptosis, and oxidative stress associated with both onset and advancement of cardiac dysfunction in T1D. Such Nrf2-mediated progression of diabetic cardiomyopathy was confirmed by a cardiomyocyte-restricted (CR) Nrf2 transgenic approach in mice. Moreover, cardiac autophagy inhibition via CR knockout of autophagy-related 5 gene (CR-Atg5KO) led to early onset and accelerated development of cardiomyopathy in T1D, and CR-Atg5KO-induced adverse phenotypes were rescued by additional Nrf2KO. Mechanistically, chronic T1D leads to glucolipotoxicity inhibiting autolysosome efflux, which in turn intensifies Nrf2-driven transcription to fuel lipid peroxidation while inactivating Nrf2-mediated antioxidant defense and impairing Nrf2-coordinated iron metabolism, thereby leading to ferroptosis in cardiomyocytes. These results demonstrate that diabetes over time causes autophagy deficiency, which turns off Nrf2-mediated defense while switching on an Nrf2-operated pathological program toward ferroptosis in cardiomyocytes, thereby worsening the progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Huimei Zang
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| | - Weiwei Wu
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| | - Lei Qi
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| | - Wenbin Tan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD
| | - Taixing Cui
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| |
Collapse
|
73
|
Giacometti J, Muhvić D, Grubić-Kezele T, Nikolić M, Šoić-Vranić T, Bajek S. Olive Leaf Polyphenols (OLPs) Stimulate GLUT4 Expression and Translocation in the Skeletal Muscle of Diabetic Rats. Int J Mol Sci 2020; 21:ijms21238981. [PMID: 33256066 PMCID: PMC7729747 DOI: 10.3390/ijms21238981] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscles are high-insulin tissues responsible for disposing of glucose via the highly regulated process of facilitated glucose transporter 4 (GLUT4). Impaired insulin action in diabetes, as well as disorders of GLUT4 vesicle trafficking in the muscle, are involved in defects in insulin-stimulated GLUT4 translocation. Since the Rab GTPases are the main regulators of vesicular membrane transport in exo- and endo-cytosis, in the present work, we studied the effect of olive leaf polyphenols (OLPs) on Rab8A, Rab13, and Rab14 proteins of the rat soleus muscle in a model of streptozotocin (SZT)-induced diabetes (DM) in a dose-dependent manner. Glucose, cholesterol, and triglyceride levels were determined in the blood, morphological changes of the muscle tissue were captured by hematoxylin and eosin histological staining, and expression of GLUT4, Rab8A, Rab13, and Rab14 proteins were analyzed in the rat soleus muscle by the immunofluorescence staining and immunoblotting. OLPs significantly reduced blood glucose level in all treated groups. Furthermore, significantly reduced blood triglycerides were found in the groups with the lowest and highest OLPs treatment. The dynamics of activation of Rab8A, Rab13, and Rab14 was OLPs dose-dependent and more effective at higher OLP doses. Thus, these results indicate a beneficial role of phenolic compounds from the olive leaf in the regulation of glucose homeostasis in the skeletal muscle.
Collapse
Affiliation(s)
- Jasminka Giacometti
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia
- Correspondence: ; Tel.: +385-51-584-557
| | - Damir Muhvić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (D.M.); (T.G.-K.)
| | - Tanja Grubić-Kezele
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (D.M.); (T.G.-K.)
- Clinical Department for Clinical Microbiology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia
| | - Marina Nikolić
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (M.N.); (T.Š.-V.); (S.B.)
| | - Tamara Šoić-Vranić
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (M.N.); (T.Š.-V.); (S.B.)
| | - Snježana Bajek
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (M.N.); (T.Š.-V.); (S.B.)
| |
Collapse
|
74
|
Meng T, Qin W, Liu B. SIRT1 Antagonizes Oxidative Stress in Diabetic Vascular Complication. Front Endocrinol (Lausanne) 2020; 11:568861. [PMID: 33304318 PMCID: PMC7701141 DOI: 10.3389/fendo.2020.568861] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022] Open
Abstract
Diabetic mellitus (DM) is a significant public health concern worldwide with an increased incidence of morbidity and mortality, which is particularly due to the diabetic vascular complications. Several pivotal underlying mechanisms are associated with vascular complications, including hyperglycemia, mitochondrial dysfunction, inflammation, and most importantly, oxidative stress. Oxidative stress triggers defective angiogenesis, activates pro-inflammatory pathways and causes long-lasting epigenetic changes to facilitate the development of vascular complications. Therefore, therapeutic interventions targeting oxidative stress are promising to manage diabetic vascular complications. Sirtuin1 (SIRT1), a class III histone deacetylase belonging to the sirtuin family, plays critical roles in regulating metabolism and ageing-related pathological conditions, such as vascular diseases. Growing evidence has indicated that SIRT1 acts as a sensing regulator in response to oxidative stress and attenuates vascular dysfunction via cooperating with adenosine-monophosphate-activated protein kinase (AMPK) to activate antioxidant signals through various downstream effectors, including peroxisome proliferator-activated receptor-gamma co-activator 1 (PGC-1α), forkhead transcription factors (FOXOs), and peroxisome proliferative-activated receptor α (PPARα). In addition, SIRT1 interacts with hydrogen sulfide (H2S), regulates NADPH oxidase, endothelial NO synthase, and mechanistic target of rapamycin (mTOR) to suppress oxidative stress. Furthermore, mRNA expression of sirt1 is affected by microRNAs in DM. In the current review, we summarize recent advances illustrating the importance of SIRT1 in antagonizing oxidative stress. We also discuss whether modulation of SIRT1 can serve as a therapeutic strategy to treat diabetic vascular complications.
Collapse
Affiliation(s)
- Teng Meng
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Weifeng Qin
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
75
|
Cardioprotective Effects of Taurisolo® in Cardiomyoblast H9c2 Cells under High-Glucose and Trimethylamine N-Oxide Treatment via De Novo Sphingolipid Synthesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2961406. [PMID: 33273998 PMCID: PMC7683148 DOI: 10.1155/2020/2961406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/21/2020] [Accepted: 10/24/2020] [Indexed: 12/27/2022]
Abstract
In addition to high plasma glucose, increased levels of trimethylamine N-oxide (TMAO) have been found in obese subjects, where are considered as a novel risk factor for cardiovascular diseases. The present study aimed to investigate the effect of a novel nutraceutical formulation based on grape polyphenols (registered as Taurisolo®) in counteracting TMAO- and high glucose (HG)-induced cytotoxicity in cardiomyoblast H9c2 cells. Cell damage was induced with HG (HG-H9c2) and HG+TMAO (THG-H9c2); both experimental cell models were, thus, incubated for 72 h in the presence or absence of Taurisolo®. It was observed that Taurisolo® significantly increased the cell viability and reduced lactate dehydrogenase and aspartate transaminase release in both HG- and THG-H9c2 cells. Additionally, through its antioxidant activity, Taurisolo® modulated cell proliferation via ERK activation in THG-H9c2. Furthermore, Taurisolo® was able to induce autophagic process via increasing the expression of LC3II, a protein marker involved in formation of autophagosome and ex novo synthesis of sphingomyelin, ceramides, and their metabolites both in HG- and THG-H9c2 cells. Finally, Taurisolo® reduced hypertrophy and induced differentiation of HG-H9C2 cells into cardiomyocyte-like cells. These data suggest that Taurisolo® counteracts the toxicity induced by TMAO and HG concentrations increasing autophagic process and activating de novo sphingolipid synthesis, resulting in a morphological cell remodeling. In conclusion, our results allow speculating that Taurisolo®, combined with energy restriction, may represent a useful nutraceutical approach for prevention of cardiomyopathy in obese subjects.
Collapse
|
76
|
Limanaqi F, Busceti CL, Biagioni F, Lazzeri G, Forte M, Schiavon S, Sciarretta S, Frati G, Fornai F. Cell Clearing Systems as Targets of Polyphenols in Viral Infections: Potential Implications for COVID-19 Pathogenesis. Antioxidants (Basel) 2020; 9:E1105. [PMID: 33182802 PMCID: PMC7697279 DOI: 10.3390/antiox9111105] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/29/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has generated the ongoing coronavirus disease-2019 (COVID-19) pandemic, still with an uncertain outcome. Besides pneumonia and acute lung injury (ALI) or acute respiratory distress syndrome (ARDS), other features became evident in the context of COVID-19. These includes endothelial and coagulation dysfunction with disseminated intravascular coagulation (DIC), and multiple organ dysfunction syndrome (MODS), along with the occurrence of neurological alterations. The multi-system nature of such viral infection is a witness to the exploitation and impairment of ubiquitous subcellular and metabolic pathways for the sake of its life-cycle, ranging from host cell invasion, replication, transmission, up to a cytopathic effect and overt systemic inflammation. In this frame, alterations in cell-clearing systems of the host are emerging as a hallmark in the pathogenesis of various respiratory viruses, including SARS-CoV-2. Indeed, exploitation of the autophagy and proteasome pathways might contribute not only to the replication of the virus at the site of infection but also to the spreading of either mature virions or inflammatory mediators at both cellular and multisystem levels. In this frame, besides a pharmacological therapy, many researchers are wondering if some non-pharmacological substances might counteract or positively modulate the course of the infection. The pharmacological properties of natural compounds have gained increasing attention in the field of alternative and adjunct therapeutic approaches to several diseases. In particular, several naturally-occurring herbal compounds (mostly polyphenols) are reported to produce widespread antiviral, anti-inflammatory, and anti-oxidant effects while acting as autophagy and (immuno)-proteasome modulators. This article attempts to bridge the perturbation of autophagy and proteasome pathways with the potentially beneficial effects of specific phytochemicals and flavonoids in viral infections, with a focus on the multisystem SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (G.L.)
| | - Carla Letizia Busceti
- I.R.C.C.S. Neuromed Pozzilli, Via Atinense, 18, 86077 Pozzilli, Italy (F.B.); (M.F.); (S.S.); (G.F.)
| | - Francesca Biagioni
- I.R.C.C.S. Neuromed Pozzilli, Via Atinense, 18, 86077 Pozzilli, Italy (F.B.); (M.F.); (S.S.); (G.F.)
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (G.L.)
| | - Maurizio Forte
- I.R.C.C.S. Neuromed Pozzilli, Via Atinense, 18, 86077 Pozzilli, Italy (F.B.); (M.F.); (S.S.); (G.F.)
| | - Sonia Schiavon
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 40100 Latina, Italy;
| | - Sebastiano Sciarretta
- I.R.C.C.S. Neuromed Pozzilli, Via Atinense, 18, 86077 Pozzilli, Italy (F.B.); (M.F.); (S.S.); (G.F.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 40100 Latina, Italy;
| | - Giacomo Frati
- I.R.C.C.S. Neuromed Pozzilli, Via Atinense, 18, 86077 Pozzilli, Italy (F.B.); (M.F.); (S.S.); (G.F.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 40100 Latina, Italy;
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (G.L.)
- I.R.C.C.S. Neuromed Pozzilli, Via Atinense, 18, 86077 Pozzilli, Italy (F.B.); (M.F.); (S.S.); (G.F.)
| |
Collapse
|
77
|
Kaludercic N, Maiuri MC, Kaushik S, Fernández ÁF, de Bruijn J, Castoldi F, Chen Y, Ito J, Mukai R, Murakawa T, Nah J, Pietrocola F, Saito T, Sebti S, Semenzato M, Tsansizi L, Sciarretta S, Madrigal-Matute J. Comprehensive autophagy evaluation in cardiac disease models. Cardiovasc Res 2020; 116:483-504. [PMID: 31504266 PMCID: PMC7064050 DOI: 10.1093/cvr/cvz233] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/01/2019] [Accepted: 08/22/2019] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a highly conserved recycling mechanism essential for maintaining cellular homeostasis. The pathophysiological role of autophagy has been explored since its discovery 50 years ago, but interest in autophagy has grown exponentially over the last years. Many researchers around the globe have found that autophagy is a critical pathway involved in the pathogenesis of cardiac diseases. Several groups have created novel and powerful tools for gaining deeper insights into the role of autophagy in the aetiology and development of pathologies affecting the heart. Here, we discuss how established and emerging methods to study autophagy can be used to unravel the precise function of this central recycling mechanism in the cardiac system.
Collapse
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, Department of Biomedical Sciences, National Research Council of Italy (CNR), 35131, Padova, Italy
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, 75006, Paris, France
| | - Susmita Kaushik
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Álvaro F Fernández
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jenny de Bruijn
- Department of Pathology, Cardiovascular Research Institute (CARIM), Maastricht University, P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands; Institute of Molecular Cardiovascular Research (IMCAR), RWTH Aachen, University, Pauwelsstrase 30, 52074, Aachen, Germany
| | - Francesca Castoldi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, 75006, Paris, France
| | - Yun Chen
- Departments of Medicine (Cardiology) and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Jumpei Ito
- The School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, UK
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NY, USA
| | - Tomokazu Murakawa
- The School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, UK
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NY, USA
| | - Federico Pietrocola
- Cellular Plasticity and Disease Laboratory. Institute for Research in Biomedicine (IRB Barcelona), Barcelona; Institute of Science and Technology (BIST), Barcelona, Spain
| | - Toshiro Saito
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Salwa Sebti
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Martina Semenzato
- Department of Biology, University of Padua, Via U Bassi 58B, 35121, Padua, Italy.,Venetian Institute of Molecular Medicine, Via Orus 2, 35129, Padua, Italy
| | - Lorenza Tsansizi
- Department of Biology, University of Padua, Via U Bassi 58B, 35121, Padua, Italy.,Venetian Institute of Molecular Medicine, Via Orus 2, 35129, Padua, Italy
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100, Latina, LT, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, 86077, Pozzilli, IS, Italy
| | - Julio Madrigal-Matute
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| |
Collapse
|
78
|
Kaur N, Raja R, Ruiz-Velasco A, Liu W. Cellular Protein Quality Control in Diabetic Cardiomyopathy: From Bench to Bedside. Front Cardiovasc Med 2020; 7:585309. [PMID: 33195472 PMCID: PMC7593653 DOI: 10.3389/fcvm.2020.585309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure is a serious comorbidity and the most common cause of mortality in diabetes patients. Diabetic cardiomyopathy (DCM) features impaired cellular structure and function, culminating in heart failure; however, there is a dearth of specific clinical therapy for treating DCM. Protein homeostasis is pivotal for the maintenance of cellular viability under physiological and pathological conditions, particularly in the irreplaceable cardiomyocytes; therefore, it is tightly regulated by a protein quality control (PQC) system. Three evolutionarily conserved molecular processes, the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), and autophagy, enhance protein turnover and preserve protein homeostasis by suppressing protein translation, degrading misfolded or unfolded proteins in cytosol or organelles, disposing of damaged and toxic proteins, recycling essential amino acids, and eliminating insoluble protein aggregates. In response to increased cellular protein demand under pathological insults, including the diabetic condition, a coordinated PQC system retains cardiac protein homeostasis and heart performance, on the contrary, inappropriate PQC function exaggerates cardiac proteotoxicity with subsequent heart dysfunction. Further investigation of the PQC mechanisms in diabetes propels a more comprehensive understanding of the molecular pathogenesis of DCM and opens new prospective treatment strategies for heart disease and heart failure in diabetes patients. In this review, the function and regulation of cardiac PQC machinery in diabetes mellitus, and the therapeutic potential for the diabetic heart are discussed.
Collapse
Affiliation(s)
- Namrita Kaur
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Rida Raja
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrea Ruiz-Velasco
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
79
|
Wu X, Liu Z, Yu XY, Xu S, Luo J. Autophagy and cardiac diseases: Therapeutic potential of natural products. Med Res Rev 2020; 41:314-341. [PMID: 32969064 DOI: 10.1002/med.21733] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/28/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022]
Abstract
The global incidence of cardiac diseases is expected to increase in the coming years, imposing a substantial socioeconomic burden on healthcare systems. Autophagy is a tightly regulated lysosomal degradation mechanism important for cell survival, homeostasis, and function. Accumulating pieces of evidence have indicated a major role of autophagy in the regulation of cardiac homeostasis and function. It is well established that dysregulation of autophagy in cardiomyocytes is involved in cardiac hypertrophy, myocardial infarction, diabetic cardiomyopathy, and heart failure. In this sense, autophagy seems to be an attractive therapeutic target for cardiac diseases. Recently, multiple natural products/phytochemicals, such as resveratrol, berberine, and curcumin have been shown to regulate cardiomyocyte autophagy via different pathways. The autophagy-modifying capacity of these compounds should be taken into consideration for designing novel therapeutic agents. This review focuses on the role of autophagy in various cardiac diseases and the pharmacological basis and therapeutic potential of reported natural products in cardiac diseases by modifying autophagic processes.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zumei Liu
- Department of Central Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Suowen Xu
- Department of Endocrinology and Metabolism, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jiandong Luo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
80
|
Packer M. Cardioprotective Effects of Sirtuin-1 and Its Downstream Effectors: Potential Role in Mediating the Heart Failure Benefits of SGLT2 (Sodium-Glucose Cotransporter 2) Inhibitors. Circ Heart Fail 2020; 13:e007197. [PMID: 32894987 DOI: 10.1161/circheartfailure.120.007197] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cardioprotective effects of SGLT2 (sodium-glucose cotransporter 2) inhibitors may be related to their ability to induce a fasting-like paradigm, which triggers the activation of nutrient deprivation pathways to promote cellular homeostasis. The most distinctive metabolic manifestations of this fasting mimicry are enhanced gluconeogenesis and ketogenesis, which are not seen with other antihyperglycemic drugs. The principal molecular stimulus to gluconeogenesis and ketogenesis is activation of SIRT1 (sirtuin-1) and its downstream mediators: PGC-1α (proliferator-activated receptor gamma coactivator 1-alpha) and FGF21 (fibroblast growth factor 21). These three nutrient deprivation sensors exert striking cardioprotective effects in a broad range of experimental models. This benefit appears to be related to their actions to alleviate oxidative stress and promote autophagy-a lysosome-dependent degradative pathway that disposes of dysfunctional organelles that are major sources of cellular injury. Nutrient deprivation sensors are suppressed in states of perceived energy surplus (ie, type 2 diabetes mellitus and chronic heart failure), but SGLT2 inhibitors activate SIRT1/PGC-1α/FGF21 signaling and promote autophagy. This effect may be related to their action to trigger the perception of a system-wide decrease in environmental nutrients, but SGLT2 inhibitors may also upregulate SIRT1, PGC-1α, and FGF21 by a direct effect on the heart. Interestingly, metformin-induced stimulation of AMP-activated protein kinase (a nutrient deprivation sensor that does not promote ketogenesis) has not been shown to reduce heart failure events in clinical trials. Therefore, promotion of ketogenic nutrient deprivation signaling by SGLT2 inhibitors may explain their cardioprotective effects, even though SGLT2 is not expressed in the heart.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX;and Imperial College, London, United Kingdom
| |
Collapse
|
81
|
Zhang D, He Y, Ye X, Cai Y, Xu J, Zhang L, Li M, Liu H, Wang S, Xia Z. Activation of autophagy inhibits nucleotide-binding oligomerization domain-like receptor protein 3 inflammasome activation and attenuates myocardial ischemia-reperfusion injury in diabetic rats. J Diabetes Investig 2020; 11:1126-1136. [PMID: 32064785 PMCID: PMC7477534 DOI: 10.1111/jdi.13235] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/16/2020] [Accepted: 02/13/2020] [Indexed: 12/29/2022] Open
Abstract
AIMS/INTRODUCTION Diabetic hearts are more vulnerable to ischemia-reperfusion injury (I/RI). The activation of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome can mediate the inflammatory process, and hence might contribute to myocardial I/RI. Activation of autophagy can eliminate excess reactive oxygen species and alleviate myocardial I/RI in diabetes. The present study aimed to investigate whether the activation of autophagy can alleviate diabetic myocardial I/RI through inhibition of NLRP3 inflammasome activation. MATERIALS AND METHODS A dose of 65 mg/kg streptozotocin was given by tail vein injection to establish a type 1 diabetes model in the rats. The left anterior descending coronary artery was ligated for 30 min followed by reperfusion for 2 h to establish a myocardial I/RI model. H9C2 cardiomyocytes were exposed to high glucose (33 mmol/L) and subjected to hypoxia-reoxygenation (6 h hypoxia followed by 4 h reoxygenation). RESULTS The diabetic rats showed significant inhibition of cardiac autophagy (decreased LC3-II/I and increased p62) that was concomitant with increased activation of NLRP3 inflammasome (increased NLRP3, apoptosis-related spots protein cleaved caspase-1, interleukin-18, interleukin-1β) and more severe myocardial I/RI (elevated creatine kinase myocardial band, lactate dehydrogenase and larger infarct size). However, administration of rapamycin, an inhibitor of the autophagy, to activate autophagy resulted in the inhibition of NLRP3 inflammasome, and finally alleviated myocardial I/RI. In vitro, high glucose inhibited autophagy, while activating NLRP3 inflammasome in H9C2 cardiomyocytes and aggravating hypoxia-reoxygenation injury, but rapamycin reversed these adverse effects of high glucose. CONCLUSION Activation of autophagy can suppress the formation of NLRP3 inflammasome, which in turn attenuates myocardial ischemia-reperfusion injury in diabetic rats.
Collapse
Affiliation(s)
- Dengwen Zhang
- Department of AnesthesiologyGuangdong Cardiovascular Institute & Guangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of AnesthesiologyThe University of Hong KongHong KongChina
| | - Yi He
- Department of AnesthesiologyGuangdong Cardiovascular Institute & Guangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xiaodong Ye
- Department of AnesthesiologyThe University of Hong KongHong KongChina
| | - Yin Cai
- Department of AnesthesiologyThe University of Hong KongHong KongChina
| | - Jindong Xu
- Department of AnesthesiologyGuangdong Cardiovascular Institute & Guangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Liangqing Zhang
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Mingyi Li
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Hao Liu
- Department of AnesthesiologyGuangzhou Medical University Second Affiliated HospitalGuangzhouChina
| | - Sheng Wang
- Department of AnesthesiologyGuangdong Cardiovascular Institute & Guangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhouChina
- Department of AnesthesiologyLinzhi People’s HospitalLinzhi, TibetChina
| | - Zhengyuan Xia
- Department of AnesthesiologyThe University of Hong KongHong KongChina
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| |
Collapse
|
82
|
A Re-Appraisal of Pathogenic Mechanisms Bridging Wet and Dry Age-Related Macular Degeneration Leads to Reconsider a Role for Phytochemicals. Int J Mol Sci 2020; 21:ijms21155563. [PMID: 32756487 PMCID: PMC7432893 DOI: 10.3390/ijms21155563] [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: 07/13/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/14/2022] Open
Abstract
Which pathogenic mechanisms underlie age-related macular degeneration (AMD)? Are they different for dry and wet variants, or do they stem from common metabolic alterations? Where shall we look for altered metabolism? Is it the inner choroid, or is it rather the choroid–retinal border? Again, since cell-clearing pathways are crucial to degrade altered proteins, which metabolic system is likely to be the most implicated, and in which cell type? Here we describe the unique clearing activity of the retinal pigment epithelium (RPE) and the relevant role of its autophagy machinery in removing altered debris, thus centering the RPE in the pathogenesis of AMD. The cell-clearing systems within the RPE may act as a kernel to regulate the redox homeostasis and the traffic of multiple proteins and organelles toward either the choroid border or the outer segments of photoreceptors. This is expected to cope with the polarity of various domains within RPE cells, with each one owning a specific metabolic activity. A defective clearance machinery may trigger unconventional solutions to avoid intracellular substrates’ accumulation through unconventional secretions. These components may be deposited between the RPE and Bruch’s membrane, thus generating the drusen, which remains the classic hallmark of AMD. These deposits may rather represent a witness of an abnormal RPE metabolism than a real pathogenic component. The empowerment of cell clearance, antioxidant, anti-inflammatory, and anti-angiogenic activity of the RPE by specific phytochemicals is here discussed.
Collapse
|
83
|
Ahmad I, Hoda M. Molecular mechanisms of action of resveratrol in modulation of diabetic and non-diabetic cardiomyopathy. Pharmacol Res 2020; 161:105112. [PMID: 32758636 DOI: 10.1016/j.phrs.2020.105112] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022]
Abstract
Cardiomyopathy is among the major clinical manifestations of heart diseases that triggers malfunctioning of the cardiovascular system. Some of the major causal factors of cardiomyopathy includes myocardial ischemia, drug-toxicity, genetic aberrations, abnormal depositions of essential elements, and redox imbalance. Diabetes, being the major comorbid of cardiovascular diseases and vice versa, further contributes to the progression of cardiomyopathy. The molecular mechanisms of action suggest that oxidative stress is among the primary factors that triggers cascading impact on cardiomyopathy. Resveratrol, a phenolic antioxidant, has the potential to quench the excessive free radicals. It is a potent antioxidant supplement that may as well be a therapeutic molecule. The review focuses on the various molecular mechanisms of action that resveratrol potentiates in reversing or attenuating the progress of diabetic and non-diabetic cardiomyopathy triggered by wide range of factors. Additionally, resveratrol also tends to preserve the healthy heart from potential damage that may be triggered by oxidative stress.
Collapse
Affiliation(s)
- Irshad Ahmad
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Muddasarul Hoda
- Department of Biological Sciences, Aliah University, IIA/27, Newtown, Kolkata, 700160, India.
| |
Collapse
|
84
|
Ying Y, Jiang C, Zhang M, Jin J, Ge S, Wang X. Phloretin protects against cardiac damage and remodeling via restoring SIRT1 and anti-inflammatory effects in the streptozotocin-induced diabetic mouse model. Aging (Albany NY) 2020; 11:2822-2835. [PMID: 31076562 PMCID: PMC6535073 DOI: 10.18632/aging.101954] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/03/2019] [Indexed: 12/29/2022]
Abstract
Diabetic cardiomyopathy increases the risk of heart failure independent of coronary artery disease and hypertension. Phloretin (PHL) shows anti-inflammatory effects in macrophages. In this study, we explored the protective effects of PHL on high glucose (HG)-induced injury in diabetic cardiomyopathy in vivo and in vitro. Using streptozotocin-induced diabetic mouse model and incubating cardiac cells line under a HG environment, PHL were evaluated of the activities of anti-inflammation and anti-fibrosis. In the study, PHL treatment ameliorated cardiomyocyte inflammation injury, and reduced fibrosis in vivo and in vitro. PHL also improved cardiac biochemical criterions after 8 weeks of induction of diabetes in C57BL/6 mice. Molecular docking results indicated that silent information regulator 2 homolog 1 (SIRT1) bound to PHL directly and that SIRT1 expression was upregulated in the PHL-treated group in HG-induced H9C2 cells. Protective effect of PHL was been eliminated in silence SIRT1 H9C2 cells. Taken together, these results suggested that PHL suppressed HG-induced cardiomyocyte injury via restoring SIRT1 expression.
Collapse
Affiliation(s)
- Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzho, Zhejiang 310012, China
| | - Cheng Jiang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzho, Zhejiang 310012, China
| | - Meiling Zhang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzho, Zhejiang 310012, China
| | - Jiye Jin
- Department of Rehabilitation, Tongde Hospital of Zhejiang Province, Hangzho, Zhejiang 310012, China
| | - Shuyu Ge
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzho, Zhejiang 310012, China
| | - Xiaodong Wang
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzho, Zhejiang 310012, China
| |
Collapse
|
85
|
Zhang J, Ren J, Liu Y, Huang D, Lu L. Resveratrol regulates the recovery of rat sciatic nerve crush injury by promoting the autophagy of Schwann cells. Life Sci 2020; 256:117959. [PMID: 32531375 DOI: 10.1016/j.lfs.2020.117959] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 05/30/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
Resveratrol has the ability to promote functional recovery after sciatic nerve crush injury (SNCI), though the mechanism through which this occurs in not fully understood. Resveratrol can promote autophagy, a key process in Wallerian degeneration; thus, we hypothesized that resveratrol could promote recovery from SNCI by promoting Schwann cell autophagy and acceleration of Wallerian degeneration. Motor function recovery was assessed by calculating Sciatic Function Indexes (SFIs) at days 7, 14, 21, 28 post SNCI. Autophagy and myelin clearance were assessed by microtubule-associated protein light chain 3B (LC3B) and myelin protein zero (MPZ) immunofluorescence and Western blot analysis on the fourth day after SNCI. The autophagy of Schwann cells following resveratrol administration was quantified by immunofluorescence in RSC96 cells. Immunofluorescence and Transmission electron microscopy (TEM) were also used in Resveratrol treated sciatic nerve four days post-SNCI to find LC3B positive areas and typical double membrane structures represent for autophagy. The SNCI+resveratrol (crush+Res) groups recovered faster than the SNCI+vehicles (crush+V) group. On day four, almost all of the myelin had regenerated in the crush+Res rats, while the crush+V group's myelin remained intact and the expression levels of LC3-II/I was the highest. On day 28 post-injury, both the control and crush+Res groups' myelin neurofibers reached peak numbers as did the thickness of the myelin sheath. Both in vitro and in vivo immunofluorescence showed that LC3B was colocalized with Schwann cells. This is the first study to observe that resveratrol can promote recovery from SCNI by accelerating the myelin clearance process by promoting autophagy of Schwann cells.
Collapse
Affiliation(s)
- Jiayi Zhang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jingyan Ren
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Dongxu Huang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Laijin Lu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
86
|
Packer M. Autophagy-dependent and -independent modulation of oxidative and organellar stress in the diabetic heart by glucose-lowering drugs. Cardiovasc Diabetol 2020; 19:62. [PMID: 32404204 PMCID: PMC7222526 DOI: 10.1186/s12933-020-01041-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/09/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a lysosome-dependent intracellular degradative pathway, which mediates the cellular adaptation to nutrient and oxygen depletion as well as to oxidative and endoplasmic reticulum stress. The molecular mechanisms that stimulate autophagy include the activation of energy deprivation sensors, sirtuin-1 (SIRT1) and adenosine monophosphate-activated protein kinase (AMPK). These enzymes not only promote organellar integrity directly, but they also enhance autophagic flux, which leads to the removal of dysfunctional mitochondria and peroxisomes. Type 2 diabetes is characterized by suppression of SIRT1 and AMPK signaling as well as an impairment of autophagy; these derangements contribute to an increase in oxidative stress and the development of cardiomyopathy. Antihyperglycemic drugs that signal through insulin may further suppress autophagy and worsen heart failure. In contrast, metformin and SGLT2 inhibitors activate SIRT1 and/or AMPK and promote autophagic flux to varying degrees in cardiomyocytes, which may explain their benefits in experimental cardiomyopathy. However, metformin and SGLT2 inhibitors differ meaningfully in the molecular mechanisms that underlie their effects on the heart. Whereas metformin primarily acts as an agonist of AMPK, SGLT2 inhibitors induce a fasting-like state that is accompanied by ketogenesis, a biomarker of enhanced SIRT1 signaling. Preferential SIRT1 activation may also explain the ability of SGLT2 inhibitors to stimulate erythropoiesis and reduce uric acid (a biomarker of oxidative stress)—effects that are not seen with metformin. Changes in both hematocrit and serum urate are the most important predictors of the ability of SGLT2 inhibitors to reduce the risk of cardiovascular death and hospitalization for heart failure in large-scale trials. Metformin and SGLT2 inhibitors may also differ in their ability to mitigate diabetes-related increases in intracellular sodium concentration and its adverse effects on mitochondrial functional integrity. Differences in the actions of SGLT2 inhibitors and metformin may reflect the distinctive molecular pathways that explain differences in the cardioprotective effects of these drugs.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, 621 N. Hall Street, Dallas, TX, 75226, USA. .,Imperial College, London, UK.
| |
Collapse
|
87
|
Zhang X, Li L, Zhang Q, Wei Q, Lin J, Jia J, Zhang J, Yan T, Lv Y, Jiang X, Zhang P, Song H, Zhang D, Huang Y. CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions. Front Cell Dev Biol 2020; 8:191. [PMID: 32363189 PMCID: PMC7180518 DOI: 10.3389/fcell.2020.00191] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
Induced autophagy is protective against myocardial hypoxia/ischemia (H/I) injury, but evidence regarding the extent of autophagic clearance under H/I and the molecular mechanisms that influence autophagic flux has scarcely been presented. Here, we report that CD38 knockout improved cardiac function and autophagic flux in CD38–/– mice and CD38–/– neonatal cardiomyocytes (CMs) under H/I conditions. Mechanistic studies demonstrated that overexpression of CD38 specifically downregulated the expression of Rab7 and its adaptor protein pleckstrin homology domain-containing protein family member 1 (PLEKHM1) through nicotinamide adenine dinucleotide (NAD)-dependent and non-NAD-dependent pathways, respectively. Loss of Rab7/PLEKHM1 impaired the fusion of autophagosomes and lysosomes, resulting in autophagosome accumulation in the myocardium and consequent cardiac dysfunction under H/I conditions. Thus, CD38 mediated autophagic flux blockade and cardiac dysfunction in a Rab7/PLEKHM1-dependent manner. These findings suggest a potential therapeutic strategy involving targeted suppression of CD38 expression.
Collapse
Affiliation(s)
- Xingyue Zhang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Lingfei Li
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Qinglin Wei
- Cholestatic Liver Diseases Center of the Institute of Digestive Disease, First Affiliated of Army Medical University, Chongqing, China
| | - Jiezhi Lin
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Jiezhi Jia
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Junhui Zhang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Tiantian Yan
- Military Burn Center, The 990th (159th) Hospital of the People's Liberation Army, Zhumadian, China
| | - Yanling Lv
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Xupin Jiang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Peng Zhang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Huapei Song
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,Department of Wound Repair, Institute of Wound Repair, The First Affiliated Hospital of South University of Science and Technology (Shenzhen Peoples Hospital), Shenzhen, China
| |
Collapse
|
88
|
Resveratrol and Diabetic Cardiomyopathy: Focusing on the Protective Signaling Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7051845. [PMID: 32256959 PMCID: PMC7094200 DOI: 10.1155/2020/7051845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/01/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a common cardiovascular complication of diabetic mellitus that is characterized by diastolic disorder in the early stage and clinical heart failure in the later stage. Presently, DCM is considered one of the major causes of death in diabetic patients. Resveratrol (RSV), a naturally occurring stilbene, is widely reported as a cardioprotective substance in many heart diseases. Thus far, the specific roles of RSV in DCM prevention and treatment have attracted great attention. Here, we discuss the roles of RSV in DCM by focusing its downstream targets from both in vivo and in vitro studies. Among such targets, Sirtuins 1/3 and AMP-activated kinase have been identified as key mediators that induce cardioprotection during hyperglycemia. In addition, many other signaling molecules (e.g., forkhead box-O3a and extracellular regulated protein kinases) are also regulated in the presence of RSV and exert beneficial effects such as opposing oxidative stress, inflammation, and apoptosis in cardiomyocytes exposed to high-glucose conditions. The beneficial potential of an RSV/stem cell cotherapy is also reviewed as a promising therapeutic strategy for preventing the development of DCM.
Collapse
|
89
|
Yan Y, Yang H, Xie Y, Ding Y, Kong D, Yu H. Research Progress on Alzheimer's Disease and Resveratrol. Neurochem Res 2020; 45:989-1006. [PMID: 32162143 DOI: 10.1007/s11064-020-03007-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), a common irreversible neurodegenerative disease characterized by amyloid-β plaques, neurofibrillary tangles, and changes in tau phosphorylation, is accompanied by memory loss and symptoms of cognitive dysfunction. Increases in disease incidence due to the ageing of the population have placed a great burden on society. To date, the mechanism of AD and the identities of adequate drugs for AD prevention and treatment have eluded the medical community. It has been confirmed that phytochemicals have certain neuroprotective effects against AD. For example, some progress has been made in research on the use of resveratrol, a natural polyphenolic phytochemical, for the prevention and treatment of AD in recent years. Elucidation of the pathogenesis of AD will create a solid foundation for drug treatment. In addition, research on resveratrol, including its mechanism of action, the roles of signalling pathways and its therapeutic targets, will provide new ideas for AD treatment, which is of great significance. In this review, we discuss the possible relationships between AD and the following factors: synapses, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPARs), silent information regulator 1 (SIRT1), and estrogens. We also discuss the findings of previous studies regarding these relationships in the context of AD treatment and further summarize research progress related to resveratrol treatment.
Collapse
Affiliation(s)
- Yan Yan
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huihuang Yang
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuxun Xie
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuanlin Ding
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Danli Kong
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| | - Haibing Yu
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
90
|
Cheng CK, Luo J, Lau CW, Chen Z, Tian XY, Huang Y. Pharmacological basis and new insights of resveratrol action in the cardiovascular system. Br J Pharmacol 2020; 177:1258-1277. [PMID: 31347157 PMCID: PMC7056472 DOI: 10.1111/bph.14801] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
Resveratrol (trans-3,4',5-trihydroxystilbene) belongs to the family of natural phytoalexins. Resveratrol first came to our attention in 1992, following reports of the cardioprotective effects of red wine. Thereafter, resveratrol was shown to exert antioxidant, anti-inflammatory, anti-proliferative, and angio-regulatory effects against atherosclerosis, ischaemia, and cardiomyopathy. This article critically reviews the current findings on the molecular basis of resveratrol-mediated cardiovascular benefits, summarizing the broad effects of resveratrol on longevity regulation, energy metabolism, stress resistance, exercise mimetics, circadian clock, and microbiota composition. In addition, this article also provides an update, both preclinically and clinically, on resveratrol-induced cardiovascular protection and discusses the adverse and inconsistent effects of resveratrol reported in both preclinical and clinical studies. Although resveratrol has been claimed as a master anti-aging agent against several age-associated diseases, further detailed mechanistic investigation is still required to thoroughly unravel the therapeutic value of resveratrol against cardiovascular diseases at different stages of disease development. LINKED ARTICLES: This article is part of a themed section on The Pharmacology of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.6/issuetoc.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Jiang‐Yun Luo
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Chi Wai Lau
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Zhen‐Yu Chen
- Food and Nutritional Sciences Programme, School of Life SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Xiao Yu Tian
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Yu Huang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| |
Collapse
|
91
|
The Citrus Flavonoid Naringenin Protects the Myocardium from Ageing-Dependent Dysfunction: Potential Role of SIRT1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4650207. [PMID: 32047577 PMCID: PMC7003265 DOI: 10.1155/2020/4650207] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/04/2019] [Accepted: 12/23/2019] [Indexed: 02/04/2023]
Abstract
Sirtuin 1 (SIRT1) enzyme plays a pivotal role in the regulation of many physiological functions. In particular, it is implicated in ageing-related diseases, such as cardiac hypertrophy, myocardial infarct, and endothelial dysfunction; moreover, its expression decreases with age. Therefore, an effective strategy to extend the lifespan and improve cardiovascular function is the enhancement of the expression/activity of SIRT1 with exogenous agents. The Citrus flavonoid naringenin (NAR) presents structural similarity with the natural SIRT1 activator resveratrol. In this study, we demonstrate through in vitro assays that NAR significantly activates SIRT1 enzyme and shows antisenescence effects. The binding mode of NAR into SIRT1 was detailed investigated through in silico studies. Moreover, chronic administration (for six months) of NAR (100 mg/kg/day) to 6-month-old mice leads to an enhancement of SIRT1 expression and a marked reduction of reactive oxygen species production in myocardial tissue. Furthermore, at the end of the treatment, the plasma levels of two well-known markers of cardiovascular inflammation, TNF-α and IL6, are significantly reduced in 12-month-old mice treated with NAR, as well as the cardiovascular risk (total cholesterol/HDL ratio) compared to control mice. Finally, the age-associated fibrotic remodeling, which is well detected through a Mallory trichrome staining in the vehicle-treated 12-month-old mice, is significantly reduced by the chronic treatment with NAR. Moreover, an improvement of myocardium functionality is highlighted by the enhancement of citrate synthase activity and stabilization of the mitochondrial membrane potential after NAR treatment. Taken together, these results suggest that a nutraceutical approach with NAR may have positive impacts on many critical hallmarks of myocardial senescence, contributing to improve the cardiac performance in aged subjects.
Collapse
|
92
|
Wang(a) J, Wang S, Wang(b) J, Xiao M, Guo Y, Tang Y, Zhang J, Gu J. Epigenetic Regulation Associated With Sirtuin 1 in Complications of Diabetes Mellitus. Front Endocrinol (Lausanne) 2020; 11:598012. [PMID: 33537003 PMCID: PMC7848207 DOI: 10.3389/fendo.2020.598012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 11/27/2020] [Indexed: 01/19/2023] Open
Abstract
Diabetes mellitus (DM) has been one of the largest health concerns of the 21st century due to the serious complications associated with the disease. Therefore, it is essential to investigate the pathogenesis of DM and develop novel strategies to reduce the burden of diabetic complications. Sirtuin 1 (SIRT1), a nicotinamide adenosine dinucleotide (NAD+)-dependent deacetylase, has been reported to not only deacetylate histones to modulate chromatin function but also deacetylate numerous transcription factors to regulate the expression of target genes, both positively and negatively. SIRT1 also plays a crucial role in regulating histone and DNA methylation through the recruitment of other nuclear enzymes to the chromatin. Furthermore, SIRT1 has been verified as a direct target of many microRNAs (miRNAs). Recently, numerous studies have explored the key roles of SIRT1 and other related epigenetic mechanisms in diabetic complications. Thus, this review aims to present a summary of the rapidly growing field of epigenetic regulatory mechanisms, as well as the epigenetic influence of SIRT1 on the development and progression of diabetic complications, including cardiomyopathy, nephropathy, and retinopathy.
Collapse
Affiliation(s)
- Jie Wang(a)
- School of Nursing, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shudong Wang
- Department of Cardiology at the First Hospital of Jilin University, Changchun, China
| | - Jie Wang(b)
- School of Nursing, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mengjie Xiao
- School of Nursing, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanfang Guo
- School of Nursing, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People’s Hospital of Liaoning Province, Shenyang, China
| | - Junlian Gu
- School of Nursing, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Junlian Gu,
| |
Collapse
|
93
|
Application of Autophagy in Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:265-270. [PMID: 32671754 DOI: 10.1007/978-981-15-4272-5_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Autophagy is closely related to the pathogenesis and progression of cardiovascular diseases. Autophagy may be a therapeutic target for many cardiovascular diseases. In this chapter, we will summarize autophagy activators and inhibitors as potential drugs for cardiovascular diseases.
Collapse
|
94
|
Pham DC, Shibu MA, Mahalakshmi B, Velmurugan BK. Effects of phytochemicals on cellular signaling: reviewing their recent usage approaches. Crit Rev Food Sci Nutr 2019; 60:3522-3546. [PMID: 31822111 DOI: 10.1080/10408398.2019.1699014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Most of the previous studies in last three decades report evidence of interactions between the different phytochemicals and the proteins involved in signal transduction pathways using in silico, in vitro, ex vivo, and in vivo analyses. However, extrapolation of these findings for clinical purposes has not been that fruitful. The efficacy of the phytochemicals in vivo studies is limited by parameters such as solubility, metabolic degradation, excretion, etc. Various approaches have now been devised to circumvent these limitations. Recently, chemical modification of the phytochemicals are demonstrated to reduce some of the limitations and improve their efficacy. Similar to traditional medicines several combinatorial phytochemical formulations have shown to be more efficient. Further, phytochemicals have been reported to be even more efficient in the form of nanoparticles. However, systematic evaluation of their efficacy, mode of action in pathway modulation, usage and associated challenges is required to be done. The present review begins with basic understanding of how signaling cascades regulate cellular response and the consequences of their dysregulation further summarizing the developments and problems associated with the dietary phytochemicals and also discuss recent approaches in strengthening these compounds in pharmacological applications. Only context relevant studies have been reviewed. Considering the limitations and scope of the article, authors do not claim inclusion of all the early and recent studies.
Collapse
Affiliation(s)
- Dinh-Chuong Pham
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - M A Shibu
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Bharath Kumar Velmurugan
- Toxicology and Biomedicine Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
95
|
Xu H, Cheng J, Wang X, Liu H, Wang S, Wu J, Xu B, Chen A, He F. Resveratrol pretreatment alleviates myocardial ischemia/reperfusion injury by inhibiting STIM1-mediated intracellular calcium accumulation. J Physiol Biochem 2019; 75:607-618. [DOI: 10.1007/s13105-019-00704-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022]
|
96
|
Cheng Z. The FoxO-Autophagy Axis in Health and Disease. Trends Endocrinol Metab 2019; 30:658-671. [PMID: 31443842 DOI: 10.1016/j.tem.2019.07.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 12/21/2022]
Abstract
Autophagy controls cellular remodeling and quality control. Dysregulated autophagy has been implicated in several human diseases including obesity, diabetes, cardiovascular disease, neurodegenerative diseases, and cancer. Current evidence has revealed that FoxO (forkhead box class O) transcription factors have a multifaceted role in autophagy regulation and dysregulation. Nuclear FoxOs transactivate genes that control the formation of autophagosomes and their fusion with lysosomes. Independently of transactivation, cytosolic FoxO proteins induce autophagy by directly interacting with autophagy proteins. Autophagy is also controlled by FoxOs through epigenetic mechanisms. Moreover, FoxO proteins can be degraded directly or indirectly by autophagy. Cutting-edge evidence is reviewed that the FoxO-autophagy axis plays a crucial role in health and disease.
Collapse
Affiliation(s)
- Zhiyong Cheng
- Food Science and Human Nutrition Department, The University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
97
|
Qu X, Chen X, Shi Q, Wang X, Wang D, Yang L. Resveratrol alleviates ischemia/reperfusion injury of diabetic myocardium via inducing autophagy. Exp Ther Med 2019; 18:2719-2725. [PMID: 31555372 DOI: 10.3892/etm.2019.7846] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 06/13/2019] [Indexed: 12/23/2022] Open
Abstract
The rising incidence and complications of diabetes constitutes a major public health issue. The mortality rate of diabetes-induced myocardial ischemia/reperfusion (I/R) injury is significantly elevated. Resveratrol (RSV) is a naturally occurring polyphenol considered to be a potent cardioprotective compound. The aim of the present study was to explore the function and molecular mechanism of RSV on diabetes-induced myocardial I/R injury. Left anterior descending coronary artery ligation was performed to stimulate myocardial I/R injury in streptozotocin-induced diabetic rats. Heart electrical activity was monitored through an electrocardiogram to confirm successful models. The myocardial infarct volume was detected via 2,3,5-triphenyltetrazolium chloride staining. Western blotting was employed to examine the levels of autophagy markers. It was found that the injection of RSV mitigated the ischemia- or I/R injury-induced myocardial damage on hemodynamic function and infarct size, but the autophagy inhibitor 3-methyladenine significantly blocked the function of RSV. Furthermore, the application of RSV significantly enhanced the expression of Beclin-1 and LC-3II but inhibited the serum levels of tumor necrosis factor-α and interleukin-6. These findings revealed an alleviating effect of RSV on diabetes-induced myocardial I/R injury and provided new evidence for the successful application of RSV on the diabetic myocardium.
Collapse
Affiliation(s)
- Xianfeng Qu
- Department of Anesthesiology, Municipal Hospital of Taizhou, Taizhou, Zhejiang 318000, P.R. China
| | - Xiao Chen
- Department of Anesthesiology, Municipal Hospital of Taizhou, Taizhou, Zhejiang 318000, P.R. China
| | - Qingqing Shi
- Department of Anesthesiology, Municipal Hospital of Taizhou, Taizhou, Zhejiang 318000, P.R. China
| | - Xiaofei Wang
- Department of Anesthesiology, Municipal Hospital of Taizhou, Taizhou, Zhejiang 318000, P.R. China
| | - Dongguo Wang
- Clinical Laboratory, Municipal Hospital of Taizhou, Taizhou, Zhejiang 318000, P.R. China
| | - Li Yang
- Department of Anesthesiology, Municipal Hospital of Taizhou, Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|
98
|
Zhou X, Yang J, Zhou M, Zhang Y, Liu Y, Hou P, Zeng X, Yi L, Mi M. Resveratrol attenuates endothelial oxidative injury by inducing autophagy via the activation of transcription factor EB. Nutr Metab (Lond) 2019; 16:42. [PMID: 31303889 PMCID: PMC6604179 DOI: 10.1186/s12986-019-0371-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Background Endothelial oxidative injury is a key event in the pathogenesis of atherosclerosis (AS). Resveratrol (RSV) attenuates the oxidative injury in human umbilical vein endothelial cells (HUVECs). Autophagy is critical for the RSV-induced protective effects. However, the exact underlying mechanisms haven’t been completely elucidated. Thus, we aimed to explore the role of autophagy of the anti-oxidation of RSV and the underlying mechanism in palmitic acid (PA)-stimulated HUVECs. Methods HUVECs were pretreated with 10 μM of RSV for 2 h and treated with 200 μM of PA for an additional 24 h. Cell viability, intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) levels were estimated with a microplate reader and confocal microscope. Autophagosomes were analyzed by transmission electron microscopy, while lysosomes by confocal microscopy. The expression of transcription factor EB (TFEB) and related genes were quantified by qRT-PCR assay. Furthermore, TFEB levels, autophagy, and lysosomes were examined by western blot assay. Results RSV pretreatment suppressed the PA-induced decline in cell viability and elevation in ROS and MDA levels in HUVECs. RSV pretreatment also increased LC3 production and P62 degradation while promoted the autophagosomes formation. However, 3-methyladenine (3-MA) treatment attenuated RSV-induced autophagy. RSV pretreatment upregulated the TFEB and TFEB-modulated downstream genes expression in a concentration-dependent manner. Additionally, in cells transfected with TFEB small interfering RNA, RSV-induced TFEB expression and subsequent autophagy were abolished. Meanwhile, the TFEB-modulated genes expression, the lysosomes formation and the RSV-induced anti-oxidation were suppressed. Conclusions In HUVECs, RSV attenuates endothelial oxidative injury by inducing autophagy in a TFEB-dependent manner.
Collapse
Affiliation(s)
- Xi Zhou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Jining Yang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Min Zhou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Yu Zhang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Yang Liu
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Pengfei Hou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Xianglong Zeng
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Long Yi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO.30 Gao Tan Yan Street, Shapingba District, Chongqing, 400038 People's Republic of China
| |
Collapse
|
99
|
Tan Z, Liu H, Song X, Ling Y, He S, Yan Y, Yan J, Wang S, Wang X, Chen A. Honokiol post-treatment ameliorates myocardial ischemia/reperfusion injury by enhancing autophagic flux and reducing intracellular ROS production. Chem Biol Interact 2019; 307:82-90. [DOI: 10.1016/j.cbi.2019.04.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/18/2019] [Accepted: 04/25/2019] [Indexed: 11/29/2022]
|
100
|
Wei J, Xu L, Du YN, Tang XF, Ye MQ, Wu YJ, Han WQ, Gao PJ. Membrane raft redox signalling contributes to endothelial dysfunction and vascular remodelling of thoracic aorta in angiotensin II-infused rats. Exp Physiol 2019; 104:946-956. [PMID: 30924217 DOI: 10.1113/ep087335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 03/12/2019] [Indexed: 12/21/2022]
Abstract
NEW FINDINGS What is the central question of this study? Is the membrane raft redox signalling pathway involved in blood pressure increase, endothelial dysfunction and vascular remodelling in an angiotensin II-induced hypertensive animal model? What is the main finding and its importance? The membrane raft redox signalling pathway was involved in endothelial dysfunction and medial remodelling in angiotensin II-induced hypertension. ABSTRACT The membrane raft (MR) redox pathway is characterized by NADPH oxidase activation via the clustering of its subunits through lysosome fusion and the activation of acid sphingomyelinase (ASMase). Our previous study shows that the MR redox signalling pathway is associated with angiontensin II (AngII)-induced production of reactive oxygen species (ROS) and endothelial dysfunction in rat mesenteric arteries. In the present study, we hypothesized that this signalling pathway is involved in blood pressure increase, endothelial dysfunction and vascular remodelling in an AngII-induced hypertensive animal model. Sixteen-week-old male Sprague-Dawley rats were subjected to AngII infusion for 2 weeks with or without treatment with the lysosome fusion inhibitor bafilomycin A1 and ASMase inhibitor amitriptyline. After treatments, aortas were harvested for further study. The results showed that the MR redox signalling pathway was activated as indicated by the increase of MR formation, ASMase activity and ROS production in aorta from AngII-infused rats compared with that from control rats. MR formation and ROS production were significantly inhibited in thoracic aorta from AngII-induced rats treated with bafilomycin A1 and amitriptyline. Both treatments significantly attenuated blood pressure increase, endothelial dysfunction and vascular remodelling including medial hypertrophy, and increased collagen and fibronectin deposition in thoracic aortas from AngII-infused rats. Finally, both treatments significantly prevented the increase of inflammatory factors including monocyte chemotactic protein 1, intercellular adhesion molecule 1 and tumour necrosis factor α in thoracic aorta from AngII-infused rats. In conclusion, the present study demonstrates that the MR redox signalling pathway was involved in endothelial dysfunction and medial remodelling in AngII-induced hypertension.
Collapse
Affiliation(s)
- Jian Wei
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Lian Xu
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Ya-Nan Du
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Xiao-Feng Tang
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Mao-Qing Ye
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Yong-Jie Wu
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Wei-Qing Han
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ping-Jin Gao
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|