101
|
Zhang H, Liao J, Jin L, Lin Y. NLRP3 inflammasome involves in the pathophysiology of sepsis-induced myocardial dysfunction by multiple mechanisms. Biomed Pharmacother 2023; 167:115497. [PMID: 37741253 DOI: 10.1016/j.biopha.2023.115497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is one of the serious health-affecting problems worldwide. At present, the mechanisms of SIMD are still not clearly elucidated. The NOD-like receptor protein 3 (NLRP3) inflammasome has been assumed to be involved in the pathophysiology of SIMD by regulating multiple biological processes. NLRP3 inflammasome and its related signaling pathways might affect the regulation of inflammation, autophagy, apoptosis, and pyroptosis in SIMD. A few molecular specific inhibitors of NLRP3 inflammasome (e.g., Melatonin, Ulinastatin, Irisin, Nifuroxazide, and Ginsenoside Rg1, etc.) have been developed, which showed a promising anti-inflammatory effect in a cellular or animal model of SIMD. These experimental findings indicated that NLRP3 inflammasome could be a promising therapeutic target for SIMD treatment. However, the clinical translation of NLRP3 inhibitors for treating SIMD still requires robust in vivo and preclinical trials.
Collapse
Affiliation(s)
- Hongwei Zhang
- Department of Emergency Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Jian Liao
- Department of Nephrology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang, China
| | - Litong Jin
- Department of Emergency Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Yan Lin
- Department of Critical Care Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China.
| |
Collapse
|
102
|
Zhang X, Luo Z, Li J, Lin Y, Li Y, Li W. Sestrin2 in diabetes and diabetic complications. Front Endocrinol (Lausanne) 2023; 14:1274686. [PMID: 37920252 PMCID: PMC10619741 DOI: 10.3389/fendo.2023.1274686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
Diabetes is a global health problem which is accompanied with multi-systemic complications. It is of great significance to elucidate the pathogenesis and to identify novel therapies of diabetes and diabetic complications. Sestrin2, a stress-inducible protein, is primarily involved in cellular responses to various stresses. It plays critical roles in regulating a series of cellular events, such as oxidative stress, mitochondrial function and endoplasmic reticulum stress. Researches investigating the correlations between Sestrin2, diabetes and diabetic complications are increasing in recent years. This review incorporates recent findings, demonstrates the diverse functions and regulating mechanisms of Sestrin2, and discusses the potential roles of Sestrin2 in the pathogenesis of diabetes and diabetic complications, hoping to highlight a promising therapeutic direction.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zirui Luo
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Jiahong Li
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Yaxuan Lin
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Yu Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wangen Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
103
|
Saha S, Fang X, Green CD, Das A. mTORC1 and SGLT2 Inhibitors-A Therapeutic Perspective for Diabetic Cardiomyopathy. Int J Mol Sci 2023; 24:15078. [PMID: 37894760 PMCID: PMC10606418 DOI: 10.3390/ijms242015078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic cardiomyopathy is a critical diabetes-mediated co-morbidity characterized by cardiac dysfunction and heart failure, without predisposing hypertensive or atherosclerotic conditions. Metabolic insulin resistance, promoting hyperglycemia and hyperlipidemia, is the primary cause of diabetes-related disorders, but ambiguous tissue-specific insulin sensitivity has shed light on the importance of identifying a unified target paradigm for both the glycemic and non-glycemic context of type 2 diabetes (T2D). Several studies have indicated hyperactivation of the mammalian target of rapamycin (mTOR), specifically complex 1 (mTORC1), as a critical mediator of T2D pathophysiology by promoting insulin resistance, hyperlipidemia, inflammation, vasoconstriction, and stress. Moreover, mTORC1 inhibitors like rapamycin and their analogs have shown significant benefits in diabetes and related cardiac dysfunction. Recently, FDA-approved anti-hyperglycemic sodium-glucose co-transporter 2 inhibitors (SGLT2is) have gained therapeutic popularity for T2D and diabetic cardiomyopathy, even acknowledging the absence of SGLT2 channels in the heart. Recent studies have proposed SGLT2-independent drug mechanisms to ascertain their cardioprotective benefits by regulating sodium homeostasis and mimicking energy deprivation. In this review, we systematically discuss the role of mTORC1 as a unified, eminent target to treat T2D-mediated cardiac dysfunction and scrutinize whether SGLT2is can target mTORC1 signaling to benefit patients with diabetic cardiomyopathy. Further studies are warranted to establish the underlying cardioprotective mechanisms of SGLT2is under diabetic conditions, with selective inhibition of cardiac mTORC1 but the concomitant activation of mTORC2 (mTOR complex 2) signaling.
Collapse
Affiliation(s)
- Sumit Saha
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Christopher D. Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
104
|
Gao Y, Liu J, Li K, Li T, Li R, Zhang W, Zhang X, Wang Y, Chen M, Shi R, Cao J. Metformin Alleviates Sepsis-Associated Myocardial Injury by Enhancing AMP-Activated Protein Kinase/Mammalian Target of Rapamycin Signaling Pathway-Mediated Autophagy. J Cardiovasc Pharmacol 2023; 82:308-317. [PMID: 37499052 DOI: 10.1097/fjc.0000000000001463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023]
Abstract
ABSTRACT Sepsis-associated myocardial injury is one of the main causes of death in intensive care units, and current clinical treatments have not been satisfactory. Therefore, finding an effective intervention is an urgent requirement. Metformin, an anti-type 2 diabetes drug, has been reported to be an autophagic activator agent that confers protection in some diseases. However, it is unclear whether it can provide defense against sepsis-associated myocardial injury. In this study, we investigated the cardioprotective effects of metformin pretreatment against lipopolysaccharide (LPS)-induced myocardial injury in C57BL/6J mice or H9c2 cells and the possible underlying mechanisms. Metformin was administered at a dose of 100 mg/kg for a week before LPS intraperitoneal injection. Twenty-four hours after LPS intervention, echocardiographic evaluation, reactive oxygen species measurement, Hoechst staining, western blotting, hematoxylin and eosin staining, and enzyme-linked immunosorbent assay were performed. Inhibitors of autophagy and AMP-activated protein kinase (AMPK) were used to further clarify the mechanisms involved. Metformin pretreatment effectively attenuated cardiac dysfunction, reduced the levels of myocardial enzymes, and alleviated cardiac hydroncus in LPS-treated mice. In addition, metformin restored the LPS-disrupted antioxidant defense and activated LPS-reduced autophagy by modulating the AMPK/mammalian target of rapamycin (AMPK/mTOR) pathway both in vivo and in vitro. The antioxidant effects of metformin on cardiomyocytes were abolished by the autophagy inhibitor 3-methyladenine (3-MA). Treatment with compound C, an AMPK inhibitor, reversed the metformin-induced autophagy in LPS-treated H9c2 cells. In conclusion, metformin pretreatment alleviates LPS-induced myocardial injury by activating AMPK/mTOR pathway-mediated autophagy.
Collapse
Affiliation(s)
- Yu Gao
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiao Liu
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China; and
| | - Kemin Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tian Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruihan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China; and
| | - Wenlong Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China; and
| | - Xuanping Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Min Chen
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruizan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Cao
- Department of Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
105
|
Lee HS, Noh JY, Song JY, Cheong HJ, Kim WJ. Metformin reduces the risk of developing influenza A virus related cardiovascular disease. Heliyon 2023; 9:e20284. [PMID: 37810823 PMCID: PMC10556598 DOI: 10.1016/j.heliyon.2023.e20284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023] Open
Abstract
This study investigated the drug repositioning potential of metformin for cardiovascular risk due to influenza A virus infection. Statistical analysis was performed to analyze factors related to the risk of death after IAV infection in diabetic patients. Through in vitro and in vivo experiments, the effect of metformin on influenza A virus infection in non-diabetic conditions was analyzed. In logistic regression analysis, influenza vaccination (OR = 0.378, p-value = 0.007, 0.186 < 95% C·I < 0.768) and metformin treatment (OR = 0.380, p-value = 0.016, 0.173 < 95% C·I < 0.835) were associated with a decreased influenza-related mortality in diabetic patients. In vitro and in vivo studies showed that viral replication and influenza A virus-induced cytokine expression were inhibited by metformin. In particular, MCP-1 and IP-10, cytokines related to cell infiltration and cardiovascular disease development, were significantly reduced by metformin under influenza A virus infection condition. As a result, the acute exacerbation of atherosclerosis caused by influenza A virus in mouse aorta was inhibited by metformin. In addition, we found that regulation of AKT/MAPK signaling plays an important role in the mechanism of metformin. In conclusion, we demonstrated the potential and mechanism of metformin as a treatment for acute exacerbation of atherosclerosis caused by influenza A virus infection.
Collapse
Affiliation(s)
- Han Sol Lee
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo Joo Kim
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
106
|
Golpasandi H, Rahimi MR, Ahmadi S, Łubkowska B, Cięszczyk P. Effects of Vitamin D3 Supplementation and Aerobic Training on Autophagy Signaling Proteins in a Rat Model Type 2 Diabetes Induced by High-Fat Diet and Streptozotocin. Nutrients 2023; 15:4024. [PMID: 37764807 PMCID: PMC10535215 DOI: 10.3390/nu15184024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of this study was to investigate the combined effects of vitamin D3 supplementation and aerobic training on regulating the autophagy process in rats with type 2 diabetic induced by a high-fat diet and streptozotocin. A total of 40 Wistar rats were divided into five groups: normal control (NC), diabetic control (DC), diabetic + aerobic training (DAT), diabetic + vitamin D3 (DVD), and diabetic + aerobic training + vitamin D3 (DVDAT). The rats underwent eight weeks of aerobic training with an intensity of 60% maximum running speed for one hour, along with weekly subcutaneous injections of 10,000 units of vitamin D3. The protein levels of different autophagy markers were assessed in the left ventricular heart tissue. The results showed that the protein levels of AMPK, pAMPK, mTOR, and pmTOR were significantly lower in the DC group compared to the NC group. Conversely, the levels of ULK, Beclin-1, LC3II, Fyco, and Cathepsin D proteins were significantly higher in the DC group. However, the interventions of aerobic training and vitamin D3 supplementation, either individually or in combination, led to increased levels of AMPK, pAMPK, mTOR, and pmTOR, and decreased levels of ULK, Beclin-1, LC3II, Fyco, and Cathepsin D (p < 0.05). Additionally, the aerobic capacity in the DAT and DVDAT groups was significantly higher compared to the NC, DC, and DVD groups (p < 0.05). These findings suggest that type 2 diabetes is associated with excessive autophagy in the left ventricle. However, after eight weeks of vitamin D3 supplementation and aerobic training, a significant reduction in excessive autophagy was observed in rats with type 2 diabetes.
Collapse
Affiliation(s)
- Hadi Golpasandi
- Department of Exercise Physiology, University of Kurdistan, Sanandaj 66177-15175, Iran;
| | | | - Slahadin Ahmadi
- Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj 66186-34683, Iran;
| | - Beata Łubkowska
- Faculty of Health and Life Sciences, Gdansk University of Physical Education and Sport, Gorskiego 1, 80-336 Gdansk, Poland; (B.Ł.); (P.C.)
| | - Paweł Cięszczyk
- Faculty of Health and Life Sciences, Gdansk University of Physical Education and Sport, Gorskiego 1, 80-336 Gdansk, Poland; (B.Ł.); (P.C.)
| |
Collapse
|
107
|
Nițulescu IM, Ciulei G, Cozma A, Procopciuc LM, Orășan OH. From Innate Immunity to Metabolic Disorder: A Review of the NLRP3 Inflammasome in Diabetes Mellitus. J Clin Med 2023; 12:6022. [PMID: 37762961 PMCID: PMC10531881 DOI: 10.3390/jcm12186022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
The role of the NLRP3 inflammasome is pivotal in the pathophysiology and progression of diabetes mellitus (DM), encompassing both type 1 (T1D), or type 2 (T2D). As part of the innate immune system, NLRP3 is also responsible for the chronic inflammation triggered by hyperglycemia. In both conditions, NLRP3 facilitates the release of interleukin-1β and interleukin-18. For T1D, NLRP3 perpetuates the autoimmune cascade, leading to the destruction of pancreatic islet cells. In T2D, its activation is associated with the presence of insulin resistance. NLRP3 activation is also instrumental for the presence of numerous complications associated with DM, microvascular and macrovascular. A considerable number of anti-diabetic drugs have demonstrated the ability to inhibit the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Iris Maria Nițulescu
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| | - George Ciulei
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| | - Angela Cozma
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| | - Lucia Maria Procopciuc
- Department 2 of Molecular Sciences, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Olga Hilda Orășan
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| |
Collapse
|
108
|
Lao A, Wu J, Li D, Shen A, Li Y, Zhuang Y, Lin K, Wu J, Liu J. Functionalized Metal-Organic Framework-Modified Hydrogel That Breaks the Vicious Cycle of Inflammation and ROS for Repairing of Diabetic Bone Defects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206919. [PMID: 37183293 DOI: 10.1002/smll.202206919] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/24/2023] [Indexed: 05/16/2023]
Abstract
The regeneration of diabetic bone defects remains challenging. Hyperglycemia causes inflammation state and excessive reactive oxygen species (ROS) during bone regeneration period. These two effects reinforce one another and create an endless loop that is also accompanied by mitochondrial dysfunction. However, there is still no effective and inclusive method targeting at the two aspects and breaking the vicious cycle. Herein, nanoparticles-Met@ZIF-8(metformin loaded zeolitic imidazolate frameworks) modified hydrogel that is capable of releasing metformin and Zn elements are constructed. This hydrogel treats hyperglycemia while also controlling mitochondrial function, reducing inflammation, and restoring homeostasis. In addition, the synergetic effect from metformin and Zn ions inhibits ROS-inflammation cascade generation and destroys the continuous progress by taking effects in both ROS and inflammation and further keeping organelles' homeostasis. Furthermore, with the recovery of mitochondria and breakdown of the ROS-inflammation cascade cycle, osteogenesis under a diabetic microenvironment is enhanced in vivo and in vitro. In conclusion, the study provides critical insight into the biological mechanism and potential therapy for diabetic bone regeneration.
Collapse
Affiliation(s)
- An Lao
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
- Department of Stomatology, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiaqing Wu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Dejian Li
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201301, China
| | - Aili Shen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Yaxin Li
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Yu Zhuang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Kaili Lin
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Jianyong Wu
- Department of Stomatology, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiaqiang Liu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| |
Collapse
|
109
|
Lu QB, Ding Y, Liu Y, Wang ZC, Wu YJ, Niu KM, Li KX, Zhang JR, Sun HJ. Metrnl ameliorates diabetic cardiomyopathy via inactivation of cGAS/STING signaling dependent on LKB1/AMPK/ULK1-mediated autophagy. J Adv Res 2023; 51:161-179. [PMID: 36334887 PMCID: PMC10491969 DOI: 10.1016/j.jare.2022.10.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Meteorin-like hormone (Metrnl) is ubiquitously expressed in skeletal muscle, heart, and adipose with beneficial roles in obesity, insulin resistance, and inflammation. Metrnl is found to protect against cardiac hypertrophy and doxorubicin-induced cardiotoxicity. However, its role in diabetic cardiomyopathy (DCM) is undefined. OBJECTIVES We aimed to elucidate the potential roles of Metrnl in DCM. METHODS Gain- andloss-of-function experimentswere utilized to determine the roles of Metrnl in the pathological processes of DCM. RESULTS We found that plasma Metrnl levels, myocardial Metrnl protein and mRNA expressions were significantly downregulated in both streptozotocin (STZ)-induced (T1D) mice and leptin receptor deficiency (db/db) (T2D) mice. Cardiac-specific overexpression (OE) of Metrnl markedly ameliorated cardiac injury and dysfunction in both T1D and T2D mice. In sharp contrast, specific deletion of Metrnl in the heart had the opposite phenotypes. In parallel, Metrnl OE ameliorated, whereas Metrnl downregulation exacerbated high glucose (HG)-elicited hypertrophy, apoptosis and oxidative damage in primary neonatal rat cardiomyocytes. Antibody-induced blockade of Metrnl eliminated the effects of benefits of Metrnl in vitro and in vivo. Mechanistically, Metrnl activated the autophagy pathway and inhibited the cGAS/STING signaling in a LKB1/AMPK/ULK1-dependent mechanism in cardiomyocytes. Besides, Metrnl-induced ULK1 phosphorylation facilitated the dephosphorylation and mitochondrial translocation of STING where it interacted with tumor necrosis factor receptor-associated factor 2 (TRAF2), a scaffold protein and E3 ubiquitin ligase that was responsible for ubiquitination and degradation of STING, rendering cardiomyocytes sensitive to autophagy activation. CONCLUSION Thus, Metrnl may be an attractive therapeutic target or regimen for treating DCM.
Collapse
Affiliation(s)
- Qing-Bo Lu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; Department of Endocrine, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214125, China
| | - Yi Ding
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214125, China
| | - Yao Liu
- Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zi-Chao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yu-Jie Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Kai-Ming Niu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ke-Xue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Ji-Ru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214125, China.
| | - Hai-Jian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore.
| |
Collapse
|
110
|
Zhang K, Wang T, Sun GF, Xiao JX, Jiang LP, Tou FF, Qu XH, Han XJ. Metformin protects against retinal ischemia/reperfusion injury through AMPK-mediated mitochondrial fusion. Free Radic Biol Med 2023; 205:47-61. [PMID: 37253410 DOI: 10.1016/j.freeradbiomed.2023.05.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 06/01/2023]
Abstract
Retinal ischemia/reperfusion (I/R) injury is a common pathological process responsible for cellular damage in glaucoma, diabetic retinopathy and hypertensive retinopathy. Metformin is a biguanide drug that exerts strong effects on multiple diseases. This study aims to evaluate the protective effect of metformin against retinal I/R injury and its underlying mechanism. I/R induced reduction in retina thickness and cell number in ganglion cell layer, and metformin alleviated I/R-induced retinal injury. Both retinal I/R and simulated ischemia/reperfusion (SIR) in R28 cells down-regulated expression of mitochondrial fusion protein Mfn2 and OPA1, which led to mitochondrial fission. Metformin also alleviated damage in R28 cells, and reversed the alteration in Mfn2 and OPA1, mitochondrial fission and mitochondrial membrane potential (MMP) disruption-induced by I/R or SIR as well. Intriguingly, inhibition of AMPK by compound C or siRNA prevented metformin-mediated up-regulation of Mfn2 and OPA1. Compound C and knockdown of Mfn2 or OPA1 dramatically alleviated the protective effect of metformin against intracellular ROS generation, MMP disruption, mitochondrial fission and loss of RGCs in ganglion cell layer induced by SIR or I/R. Moreover, scavenging mitochondrial ROS (mito-ROS) by mito-TEMPO exerted the similar protection against I/R-induced retinal injury or SIR-induced damage in R28 cells as metformin. Our data show for the first time that metformin protects against retinal I/R injury through AMPK-mediated mitochondrial fusion and the decreased mito-ROS generation. These findings might also repurpose metformin as a therapeutic agent for retinal I/R injury.
Collapse
Affiliation(s)
- Kun Zhang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China; Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China; Department of Ophthalmology, Shenzhen People's Hospital & the Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, 518020, PR China
| | - Tao Wang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Gui-Feng Sun
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jin-Xing Xiao
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Li-Ping Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Fang-Fang Tou
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Xin-Hui Qu
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China; The Second Department of Neurology, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China.
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China; Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China; The Second Department of Neurology, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
111
|
Bielawska M, Warszyńska M, Stefańska M, Błyszczuk P. Autophagy in Heart Failure: Insights into Mechanisms and Therapeutic Implications. J Cardiovasc Dev Dis 2023; 10:352. [PMID: 37623365 PMCID: PMC10456056 DOI: 10.3390/jcdd10080352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Autophagy, a dynamic and complex process responsible for the clearance of damaged cellular components, plays a crucial role in maintaining myocardial homeostasis. In the context of heart failure, autophagy has been recognized as a response mechanism aimed at counteracting pathogenic processes and promoting cellular health. Its relevance has been underscored not only in various animal models, but also in the human heart. Extensive research efforts have been dedicated to understanding the significance of autophagy and unravelling its complex molecular mechanisms. This review aims to consolidate the current knowledge of the involvement of autophagy during the progression of heart failure. Specifically, we provide a comprehensive overview of published data on the impact of autophagy deregulation achieved by genetic modifications or by pharmacological interventions in ischemic and non-ischemic models of heart failure. Furthermore, we delve into the intricate molecular mechanisms through which autophagy regulates crucial cellular processes within the three predominant cell populations of the heart: cardiomyocytes, cardiac fibroblasts, and endothelial cells. Finally, we emphasize the need for future research to unravel the therapeutic potential associated with targeting autophagy in the management of heart failure.
Collapse
Affiliation(s)
- Magdalena Bielawska
- Department of Clinical Immunology, Jagiellonian University Medical College, University Children’s Hospital, Wielicka 265, 30-663 Cracow, Poland; (M.B.)
| | - Marta Warszyńska
- Department of Clinical Immunology, Jagiellonian University Medical College, University Children’s Hospital, Wielicka 265, 30-663 Cracow, Poland; (M.B.)
| | - Monika Stefańska
- Department of Clinical Immunology, Jagiellonian University Medical College, University Children’s Hospital, Wielicka 265, 30-663 Cracow, Poland; (M.B.)
| | - Przemysław Błyszczuk
- Department of Clinical Immunology, Jagiellonian University Medical College, University Children’s Hospital, Wielicka 265, 30-663 Cracow, Poland; (M.B.)
- Department of Rheumatology, University Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
| |
Collapse
|
112
|
Huo JL, Feng Q, Pan S, Fu WJ, Liu Z, Liu Z. Diabetic cardiomyopathy: Early diagnostic biomarkers, pathogenetic mechanisms, and therapeutic interventions. Cell Death Discov 2023; 9:256. [PMID: 37479697 PMCID: PMC10362058 DOI: 10.1038/s41420-023-01553-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) mainly refers to myocardial metabolic dysfunction caused by high glucose, and hyperglycemia is an independent risk factor for cardiac function in the absence of coronary atherosclerosis and hypertension. DCM, which is a severe complication of diabetes, has become the leading cause of heart failure in diabetic patients. The initial symptoms are inconspicuous, and patients gradually exhibit left ventricular dysfunction and eventually develop total heart failure, which brings a great challenge to the early diagnosis of DCM. To date, the underlying pathological mechanisms of DCM are complicated and have not been fully elucidated. Although there are therapeutic strategies available for DCM, the treatment is mainly focused on controlling blood glucose and blood lipids, and there is a lack of effective drugs targeting myocardial injury. Thus, a large percentage of patients with DCM inevitably develop heart failure. Given the neglected initial symptoms, the intricate cellular and molecular mechanisms, and the lack of available drugs, it is necessary to explore early diagnostic biomarkers, further understand the signaling pathways involved in the pathogenesis of DCM, summarize the current therapeutic strategies, and develop new targeted interventions.
Collapse
Affiliation(s)
- Jin-Ling Huo
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Qi Feng
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Wen-Jia Fu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center For Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| | - Zhenzhen Liu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
113
|
Li X, Su X, Xia F, Qiu J, Zhang J, Wu H, Xie X, Xu M. Bibliometric and visual analysis of diabetes mellitus and pyroptosis from 2011 to 2022. Eur J Med Res 2023; 28:235. [PMID: 37443131 DOI: 10.1186/s40001-023-01175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
OBJECTIVE To visualize and analyze the published literature on diabetes mellitus and pyroptosis based on a bibliometric approach, so as to provide a comprehensive picture of the hot research directions and dynamic progress in this field. METHODS This study was based on the web of science core collection database to conduct a comprehensive search of the published literature in the field of diabetes mellitus and Pyroptosis from January 1985 to August 2022, including the published research literature in this field, as well as a visual analysis of the number of citations, year of publication, journal, author, research institution, country, and research topic. RESULTS A total of 139 literature on research related to diabetes mellitus and cellular scorch from 2011 to 2022 were retrieved, with a total of 3009 citations and a maximum of 255 citations for a single article, which had a first author Schmid-Burgk, JL The first author of this article is from Germany; among 20 publishing countries, China leads with 100 articles; among 222 publishing institutions, Harbin Medical University leads with 18 articles and 184 citations; among 980 authors, Chen, X from China tops the list of high-impact authors with 5 articles and 29 citations. Among the 98 journals, "CELL DEATH DISEASE" ranked first in both volume and high-impact journals with 4 articles and 29 citations. Among 349 keywords, "pyroptosis" ranked first with a cumulative frequency of 65 times. The cluster analysis was divided into three categories, chronic complications of diabetes mellitus and pyroptosis (67 articles), diabetes mellitus and pyroptosis (60 articles), and diabetes mellitus combined with other diseases and pyroptosis (12 articles), and the number of articles related to diabetes mellitus and its chronic complications increased rapidly from 2019, among which, diabetic cardiomyopathy (27 articles) had the highest number of articles. CONCLUSIONS Based on a comprehensive analysis of published literature in the field of diabetes mellitus and pyroptosis from 2011 to 2022, this study achieved a visual analysis of studies with significant and outstanding contributions to the field, thus framing a picture showing the development and changes in the field. At the same time, this study provides research information and direction for clinicians and investigators to conduct diabetes mellitus and pyroptosis-related research in the future.
Collapse
Affiliation(s)
- Xiaodong Li
- The First Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, 550000, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Xiaojuan Su
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Fenglin Xia
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jing Qiu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jiaqi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Haiyan Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xuejun Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Mingchao Xu
- Traditional Chinese Medicine Hospital of Meishan, Meishan, 620010, China.
| |
Collapse
|
114
|
Gao L, Liu X, Luo X, Lou X, Li P, Li X, Liu X. Antiaging effects of dietary supplements and natural products. Front Pharmacol 2023; 14:1192714. [PMID: 37441528 PMCID: PMC10333707 DOI: 10.3389/fphar.2023.1192714] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Aging is an inevitable process influenced by genetics, lifestyles, and environments. With the rapid social and economic development in recent decades, the proportion of the elderly has increased rapidly worldwide, and many aging-related diseases have shown an upward trend, including nervous system diseases, cardiovascular diseases, metabolic diseases, and cancer. The rising burden of aging-related diseases has become an urgent global health challenge and requires immediate attention and solutions. Natural products have been used for a long time to treat various human diseases. The primary cellular pathways that mediate the longevity-extending effects of natural products involve nutrient-sensing pathways. Among them, the sirtuin, AMP-activated protein kinase, mammalian target of rapamycin, p53, and insulin/insulin-like growth factor-1 signaling pathways are most widely studied. Several studies have reviewed the effects of individual natural compounds on aging and aging-related diseases along with the underlying mechanisms. Natural products from food sources, such as polyphenols, saponins, alkaloids, and polysaccharides, are classified as antiaging compounds that promote health and prolong life via various mechanisms. In this article, we have reviewed several recently identified natural products with potential antiaging properties and have highlighted their cellular and molecular mechanisms. The discovery and use of dietary supplements and natural products that can prevent and treat multiple aging-related diseases in humans will be beneficial. Thus, this review provides theoretical background for existing dietary supplements and natural products as potential antiaging agents.
Collapse
|
115
|
Yan M, Liu S, Zeng W, Guo Q, Mei Y, Shao X, Su L, Liu Z, Zhang Y, Wang L, Diao H, Rong X, Guo J. The Chinese herbal medicine Fufang Zhenzhu Tiaozhi ameliorates diabetic cardiomyopathy by regulating cardiac abnormal lipid metabolism and mitochondrial dynamics in diabetic mice. Biomed Pharmacother 2023; 164:114919. [PMID: 37302318 DOI: 10.1016/j.biopha.2023.114919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/03/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is an important complication leading to the death of patients with diabetes, but there is no effective strategy for clinical treatments. Fufang Zhenzhu Tiaozhi (FTZ) is a patent medicine that is a traditional Chinese medicine compound preparation with comprehensive effects for the prevention and treatment of glycolipid metabolic diseases under the guidance of "modulating liver, starting pivot and cleaning turbidity". FTZ was proposed by Professor Guo Jiao and is used for the clinical treatment of hyperlipidemia. This study was designed to explore the regulatory mechanisms of FTZ on heart lipid metabolism dysfunction and mitochondrial dynamics disorder in mice with DCM, and it provides a theoretical basis for the myocardial protective effect of FTZ in diabetes. In this study, we demonstrated that FTZ protected heart function in DCM mice and downregulated the overexpression of free fatty acids (FFAs) uptake-related proteins cluster of differentiation 36 (CD36), fatty acid binding protein 3 (FABP3) and carnitine palmitoyl transferase 1 (CPT1). Moreover, FTZ treatment showed a regulatory effect on mitochondrial dynamics by inhibiting mitochondrial fission and promoting mitochondrial fusion. We also identified in vitro that FTZ could restore lipid metabolism-related proteins, mitochondrial dynamics-related proteins and mitochondrial energy metabolism in PA-treated cardiomyocytes. Our study indicated that FTZ improves the cardiac function of diabetic mice by attenuating the increase in fasting blood glucose levels, inhibiting the decrease in body weight, alleviating disordered lipid metabolism, and restoring mitochondrial dynamics and myocardial apoptosis in diabetic mouse hearts.
Collapse
Affiliation(s)
- Meiling Yan
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Suping Liu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Wenru Zeng
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Qiaoling Guo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Yu Mei
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Xiaoqi Shao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Liyan Su
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Zhou Liu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Yue Zhang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Lexun Wang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Hongtao Diao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Xianglu Rong
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Jiao Guo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China.
| |
Collapse
|
116
|
Horváth C, Jarabicová I, Rajtík T, Bartošová L, Ferenczyová K, Kaločayová B, Barteková M, Szobi A, Adameová A. Analysis of Signaling Pathways of Necroptotic and Pyroptotic Cell Death in the Hearts of Rats With Type 2 Diabetes Mellitus. Physiol Res 2023; 72:S23-S29. [PMID: 37294115 DOI: 10.33549/physiolres.935020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
Diabetes mellitus is known to produce various cell-damaging events and thereby underlie heart dysfunction and remodeling. However, very little is known about its inflammation-associated pathomechanisms due to necrosis-like cell death. For this purpose, we aimed to investigate signaling pathways of necroptosis and pyroptosis, known to produce plasma membrane rupture with the resultant promotion of inflammation. One-year old Zucker diabetic fatty (ZDF) rats did not exhibit significant heart dysfunction as revealed by echocardiographic measurement. On the other hand, there was a decrease in heart rate due to diabetes. Immunoblotting analysis showed that the left ventricles of ZDF rats overexpress neither the main necroptotic proteins including receptor-interacting protein kinase 3 (RIP3) and mixed lineage domain kinase-like pseudokinase (MLKL), nor the pyroptotic regulators including NLR family pyrin domain containing 3 protein (NLRP3), caspase-1, interleukin-1 beta (IL-1beta and the N-terminal gasdermin D (GSDMD-N). On the other hand, the increased activation of the RIP3 kinase due to phosphorylation was found in such hearts. In summary, we showed for the first time that the activation of cardiac RIP3 is upregulated due to disturbances in glucose metabolism which, however, did not proceed to necrosis-like cell death. These data can indicate that the activated RIP3 might also underlie other pleiotropic, non-necroptotic signaling pathways under basal conditions.
Collapse
Affiliation(s)
- C Horváth
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Sánchez ML, Valdez H, Conde M, Viaña-Mendieta P, Boccaccini AR. Polymers and Bioactive Compounds with a Macrophage Modulation Effect for the Rational Design of Hydrogels for Skin Regeneration. Pharmaceutics 2023; 15:1655. [PMID: 37376103 DOI: 10.3390/pharmaceutics15061655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/08/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The development of biomaterial platforms for dispensing reagents of interest such as antioxidants, growth factors or antibiotics based on functional hydrogels represents a biotechnological solution for many challenges that the biomedicine field is facing. In this context, in situ dosing of therapeutic components for dermatological injuries such as diabetic foot ulcers is a relatively novel strategy to improve the wound healing process. Hydrogels have shown more comfort for the treatment of wounds due to their smooth surface and moisture, as well as their structural affinity with tissues in comparison to hyperbaric oxygen therapy, ultrasound, and electromagnetic therapies, negative pressure wound therapy or skin grafts. Macrophages, one of the most important cells of the innate immune system, have been described as the key not only in relation to the host immune defense, but also in the progress of wound healing. Macrophage dysfunction in chronic wounds of diabetic patients leads to a perpetuating inflammatory environment and impairs tissue repair. Modulating the macrophage phenotype from pro-inflammatory (M1) to anti-inflammatory (M2) could be a strategy for helping to improve chronic wound healing. In this regard, a new paradigm is found in the development of advanced biomaterials capable of inducing in situ macrophage polarization to offer an approach to wound care. Such an approach opens a new direction for the development of multifunctional materials in regenerative medicine. This paper surveys emerging hydrogel materials and bioactive compounds being investigated to induce the immunomodulation of macrophages. We propose four potential functional biomaterials for wound healing applications based on novel biomaterial/bioactive compound combination that are expected to show synergistic beneficial outcomes for the local differentiation of macrophages (M1-M2) as a therapeutic strategy for chronic wound healing improvement.
Collapse
Affiliation(s)
- Mirna L Sánchez
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Hugo Valdez
- Laboratorio de Microbiología Celular e Inmunomecanismos, CINDEFI|Centro de Investigación y Desarrollo en Fermentaciones Industriales Facultad de Ciencias Exactas, La Plata B1900AJL, Argentina
| | - Micaela Conde
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina
| | - Pamela Viaña-Mendieta
- Tecnologico de Monterrey, Instituto para la Investigación en Obesidad, Monterrey 64849, Mexico
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| |
Collapse
|
118
|
Rutledge CA, Lagranha C, Chiba T, Redding K, Stolz DB, Goetzman E, Sims-Lucas S, Kaufman BA. Metformin preconditioning protects against myocardial stunning and preserves protein translation in a mouse model of cardiac arrest. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100034. [PMID: 37425219 PMCID: PMC10327679 DOI: 10.1016/j.jmccpl.2023.100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Cardiac arrest (CA) causes high mortality due to multi-system organ damage attributable to ischemia-reperfusion injury. Recent work in our group found that among diabetic patients who experienced cardiac arrest, those taking metformin had less evidence of cardiac and renal damage after cardiac arrest when compared to those not taking metformin. Based on these observations, we hypothesized that metformin's protective effects in the heart were mediated by AMPK signaling, and that AMPK signaling could be targeted as a therapeutic strategy following resuscitation from CA. The current study investigates metformin interventions on cardiac and renal outcomes in a non-diabetic CA mouse model. We found that two weeks of metformin pretreatment protects against reduced ejection fraction and reduces kidney ischemia-reperfusion injury at 24 h post-arrest. This cardiac and renal protection depends on AMPK signaling, as demonstrated by outcomes in mice pretreated with the AMPK activator AICAR or metformin plus the AMPK inhibitor compound C. At this 24-h time point, heart gene expression analysis showed that metformin pretreatment caused changes supporting autophagy, antioxidant response, and protein translation. Further investigation found associated improvements in mitochondrial structure and markers of autophagy. Notably, Western analysis indicated that protein synthesis was preserved in arrest hearts of animals pretreated with metformin. The AMPK activation-mediated preservation of protein synthesis was also observed in a hypoxia/reoxygenation cell culture model. Despite the positive impacts of pretreatment in vivo and in vitro, metformin did not preserve ejection fraction when deployed at resuscitation. Taken together, we propose that metformin's in vivo cardiac preservation occurs through AMPK activation, requires adaptation before arrest, and is associated with preserved protein translation.
Collapse
Affiliation(s)
- Cody A. Rutledge
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia Lagranha
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takuto Chiba
- Rangos Research Center, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School, Pittsburgh, PA, USA
| | - Kevin Redding
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donna B. Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Goetzman
- Division of Genetic and Genomic Medicine, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sunder Sims-Lucas
- Rangos Research Center, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School, Pittsburgh, PA, USA
| | - Brett A. Kaufman
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
119
|
Yuan H, Sui H, Li S. Diosgenin alleviates the inflammatory damage and insulin resistance in high glucose‑induced podocyte cells via the AMPK/SIRT1/NF‑κB signaling pathway. Exp Ther Med 2023; 25:259. [PMID: 37153902 PMCID: PMC10155255 DOI: 10.3892/etm.2023.11958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/22/2023] [Indexed: 05/10/2023] Open
Abstract
Diabetic nephropathy (DN) is the predominant cause of end-stage renal disease globally. Diosgenin (DSG) has been reported to play a protective role in podocyte injury in DN. The present study aimed to explore the role of DSG in DN, as well as its mechanism of action in a high glucose (HG)-induced in vitro model of DN in podocytes. Cell viability, apoptosis, inflammatory response and insulin-stimulated glucose uptake were evaluated using Cell Counting Kit-8, TUNEL, ELISA and 2-deoxy-D-glucose assay, respectively. In addition, the expression of AMP-activated protein kinase (AMPK)/sirtuin 1 (SIRT1)/NF-κB signaling-related proteins in podocyte cells was measured using western blotting. The results indicated that DSG enhanced the viability of podocytes after HG exposure, but inhibited inflammatory damage and attenuated insulin resistance. Moreover, DSG induced the activation of the AMPK/SIRT1/NF-κB signaling pathway. Furthermore, treatment with compound C, an inhibitor of AMPK, counteracted the protective effects of DSG on HG-induced podocyte cells. Therefore, DSG may be a potential therapeutic compound for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Haoyu Yuan
- Department of Endocrinology, The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Huacheng Sui
- Department of Endocrinology, The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Saimei Li
- Department of Endocrinology, The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
- Correspondence to: Dr Saimei Li, Department of Endocrinology, The First Clinical College of Guangzhou University of Chinese Medicine, 12 Airport Road, Guangzhou, Guangdong 510405, P.R. China
| |
Collapse
|
120
|
Xie Y, Lei X, Zhao G, Guo R, Cui N. mTOR in programmed cell death and its therapeutic implications. Cytokine Growth Factor Rev 2023; 71-72:66-81. [PMID: 37380596 DOI: 10.1016/j.cytogfr.2023.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Mechanistic target of rapamycin (mTOR), a highly conserved serine/threonine kinase, is involved in cellular metabolism, protein synthesis, and cell death. Programmed cell death (PCD) assists in eliminating aging, damaged, or neoplastic cells, and is indispensable for sustaining normal growth, fighting pathogenic microorganisms, and maintaining body homeostasis. mTOR has crucial functions in the intricate signaling pathway network of multiple forms of PCD. mTOR can inhibit autophagy, which is part of PCD regulation. Cell survival is affected by mTOR through autophagy to control reactive oxygen species production and the degradation of pertinent proteins. Additionally, mTOR can regulate PCD in an autophagy-independent manner by affecting the expression levels of related genes and phosphorylating proteins. Therefore, mTOR acts through both autophagy-dependent and -independent pathways to regulate PCD. It is conceivable that mTOR exerts bidirectional regulation of PCD, such as ferroptosis, according to the complexity of signaling pathway networks, but the underlying mechanisms have not been fully explained. This review summarizes the recent advances in understanding mTOR-mediated regulatory mechanisms in PCD. Rigorous investigations into PCD-related signaling pathways have provided prospective therapeutic targets that may be clinically beneficial for treating various diseases.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ran Guo
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
121
|
Zhang T, Yin X, Yu X, Shang R, Lu L, Miao J. Metformin protects fibroblasts from patients with GNE myopathy by restoring autophagic flux via an AMPK/mTOR-independent pathway. Biomed Pharmacother 2023; 164:114958. [PMID: 37263165 DOI: 10.1016/j.biopha.2023.114958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/13/2023] [Accepted: 05/27/2023] [Indexed: 06/03/2023] Open
Abstract
UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) myopathy is an autosomal recessive disease characterized by rimmed vacuoles (RVs). Previous studies have shown that metformin protects against several neuromuscular disorders. In the present study, we summarize the clinical features of three GNE patients with the p.D207V mutation. The pathogenesis of GNE myopathy is described, and the significance of metformin in this disease is observed. Skin biopsy-derived fibroblasts from patients with GNE myopathy, carrying a D207V mutation in GNE, were cultured. GNE fibroblasts and control fibroblasts were treated under normal culture conditions, serum starvation conditions, or serum starvation + metformin conditions. Histopathological and immunohistochemical analyses of muscle samples showed that autophagy was involved in the formation of RVs in the muscle of patients. Starved GNE fibroblasts showed decreased autophagy-related proteins and impaired autophagic flow (p < 0.05). The mRFP-GFP-LC3 assay showed that the fusion of autophagosomes with lysosomes was partially blocked in GNE cells. Notably, metformin treatment upregulated the expression of autophagy proteins, increased the number of autolysosomes (p < 0.001), and influenced the viability of GNE cells (p < 0.001). Furthermore, adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) and phosphorylated (p)-AMPK expression levels were upregulated in serum-starved GNE fibroblasts, while the mammalian target of rapamycin (mTOR) and p-mTOR expression levels were downregulated in both groups. Metformin treatment inhibited the AMPK-mTOR signaling pathway. Our results suggest that metformin plays a protective role in the GNE fibroblast by restoring autophagic flux and through the AMPK/mTOR-independent pathway.
Collapse
Affiliation(s)
- Tongtong Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun 130000, China
| | - Xiang Yin
- Department of Neurology, The First Hospital of Jilin University, Changchun 130000, China
| | - Xuefan Yu
- Department of Neurology, The First Hospital of Jilin University, Changchun 130000, China
| | - Ren Shang
- Department of Neurology, The First Hospital of Jilin University, Changchun 130000, China
| | - Liuzhe Lu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun 130000, China
| | - Jing Miao
- Department of Neurology, The First Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
122
|
Liu Y, Lin X, Hao Z, Yu M, Tang Y, Teng X, Sun W, Kang L. Cadmium exposure caused cardiotoxicity in common carps (Cyprinus carpio L.): miR-9-5p, oxidative stress, energetic impairment, mitochondrial division/fusion imbalance, inflammation, and autophagy. FISH & SHELLFISH IMMUNOLOGY 2023; 138:108853. [PMID: 37245677 DOI: 10.1016/j.fsi.2023.108853] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 05/30/2023]
Abstract
Cadmium (Cd), a toxic heavy metal pollutant, is a threat to human and eatable fish health. Common carps are widely cultivated and eaten by humans. However, there are no reports about Cd-damaged common carp hearts. Our experiment attempted to investigate the cardiotoxicity of Cd to common carps by establishing a common carp Cd exposure model. Our results showed that Cd injured hearts. Moreover, Cd treatment induced autophagy via miR-9-5p/Sirt1/mTOR/ULK1 pathway. Cd exposure caused oxidant/antioxidant imbalance and oxidative stress; and led to energetic impairment. Energetic impairment partook in oxidative stress-mediated autophagy through AMPK/mTOR/ULK1 pathway. Furthermore, Cd caused mitochondrial division/fusion imbalance and resulted in inflammatory injury via NF-κB-COX-2-PTGEs and NF-κB-COX-2-TNF-α pathways. Oxidative stress mediated mitochondrial division/fusion imbalance, further induced inflammation and autophagy via OPA1/NF-κB-COX-2-TNF-α-Beclin1 and OPA1/NF-κB-COX-2-TNF-α/P62 pathways under Cd treatment. Taken together, miR-9-5p, oxidative stress, energetic impairment, mitochondrial division/fusion imbalance, inflammation, and autophagy participated in the mechanism of Cd-cardiotoxicity to common carps. Our study revealed harmful effect of Cd on hearts, and provided new information for researches of environmental pollutant toxicity.
Collapse
Affiliation(s)
- Yuhao Liu
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - Xu Lin
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - Zhiyu Hao
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - Meijing Yu
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China
| | - You Tang
- Electrical and Information Engineering College, JiLin Agricultural Science and Technology University, Jilin, 132101, PR China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China.
| | - Wei Sun
- College of Animal Science and Technology, Northeast Agricultural University, NO. 600 Chang Jiang Road, Xiang Fang District, Harbin, 150030, PR China.
| | - Lu Kang
- Institute of Agricultural Quality Standards and Testing Technology, Xinjiang Academy of Agricultural Sciences, Urumqi, 830091, PR China.
| |
Collapse
|
123
|
Qi Y, Chen Z, Guo B, Liu Z, Wang L, Liu S, Xue L, Ma M, Yin Y, Li Y, Liu G. Speckle-tracking echocardiography provides sensitive measurements of subtle early alterations associated with cardiac dysfunction in T2DM rats. BMC Cardiovasc Disord 2023; 23:266. [PMID: 37217862 DOI: 10.1186/s12872-023-03239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/12/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy results in cardiac structural and functional abnormalities. Previous studies have demonstrated that inhibiting the RhoA/ROCK signalling pathway increases the injury resistance of cardiomyocytes. The early detection of cardiac structural and functional alterations may facilitate an improved understanding of the pathophysiologic progress and guide therapy. This study aimed to identify the optimal diagnostic measures for the subtle early alterations of cardiac dysfunction in type 2 diabetes mellitus (T2DM) rats. METHODS Twenty-four rat models were divided into four groups and received treatments for 4 weeks: the CON group (control rats), the DM group (T2DM rats), the DMF group (T2DM rats receiving fasudil) and the CONF group (control rats receiving fasudil) group. Left ventricular (LV) structure was quantified by histological staining and transmission electron microscopy. LV function and myocardial deformation were assessed by high-frequency echocardiography. RESULTS Treatment with fasudil, a ROCK inhibitor, significantly protected against diabetes-induced myocardial hypertrophy, fibrosis and mitochondrial dysfunction. Impaired LV performance was found in T2DM rats, as evidenced by significant reductions in the ejection fraction (EF), fractional shortening (FS) and the mitral valve (MV) E/A ratio (which decreased 26%, 34% and 20%, respectively). Fasudil failed to improve the conventional ultrasonic parameters in T2DM rats, but the myocardial deformation measured by speckle-tracking echocardiography (STE) were significantly improved (global circumferential strain, GCS: P = 0.003; GCS rate, GCSR: P = 0.021). When receiver operating characteristic (ROC) curves were used in combination with linear regression analysis, STE parameters were found to be characterized by both optimal prediction of cardiac damage [AUC (95% CI): fractional area change, FAC: 0.927 (0.744, 0.993); GCS: 0.819 (0.610, 0.945); GCSR: 0.899 (0.707, 0.984)] and stronger correlations with cardiac fibrosis (FAC: r = -0.825; GCS: r = 0.772; GCSR: r = 0.829) than conventional parameters. CONCLUSION The results suggest that STE parameters are more sensitive and specific than conventional parameters in predicting the subtle cardiac functional changes that occur in the early stage, providing new insight into the management of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yanchao Qi
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Zhiyan Chen
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Bingyan Guo
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Zhe Liu
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Lijie Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Suyun Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Lixiang Xue
- Center of Basic Medical Research, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Meifang Ma
- Department of Cardiology, Handan Central Hospital, Handan, 056008, Hebei, People's Republic of China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, 050031, Hebei, People's Republic of China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, 050031, Hebei, People's Republic of China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, People's Republic of China
| | - Yongjun Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China.
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, People's Republic of China.
| |
Collapse
|
124
|
Fang B, Liu F, Yu X, Luo J, Zhang X, Zhang T, Zhang J, Yang Y, Li X. Liraglutide alleviates myocardial ischemia‒reperfusion injury in diabetic mice. Mol Cell Endocrinol 2023; 572:111954. [PMID: 37172886 DOI: 10.1016/j.mce.2023.111954] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Diabetic patients are prone to acute myocardial infarction. Although reperfusion therapy can preserve the viability of the myocardium, it also causes fatal ischemia‒reperfusion injury. Diabetes can exacerbate myocardial ischemia‒reperfusion injury, but the mechanism is unclear. We aimed to characterize the effects of liraglutide on the prevention of ischemia‒reperfusion injury and inadequate autophagy. Liraglutide reduced the myocardial infarction area and improved cardiac function in diabetic mice. We further demonstrated that liraglutide mediated these protective effects by activating AMPK/mTOR-mediated autophagy. Liraglutide markedly increased p-AMPK levels and the LC3 II/LC3 I ratio and reduced p-mTOR levels and p62 expression. Pharmacological inhibition of mTOR increased cell viability and autophagy levels in high glucose and H/R-treated H9C2 cells. Overall, our study reveals that liraglutide acts upstream of the AMPK/mTOR pathway to effectively counteract high glucose- and H/R-induced cell dysfunction by activating AMPK/mTOR-dependent autophagy, providing a basis for the clinical prevention and treatment of ischemia‒reperfusion in diabetes.
Collapse
Affiliation(s)
- Binbin Fang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Fen Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaolin Yu
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Junyi Luo
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xuehe Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tong Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jixin Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yining Yang
- Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Xiaomei Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
125
|
Xu Y, Cao L, Zou W, Yu R, Shen W. Panax notoginseng saponins inhibits NLRP3 inflammasome-mediated pyroptosis by downregulating lncRNA-ANRIL in cardiorenal syndrome type 4. Chin Med 2023; 18:50. [PMID: 37158944 PMCID: PMC10165771 DOI: 10.1186/s13020-023-00756-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/24/2023] [Indexed: 05/10/2023] Open
Abstract
OBJECTIVE Cardiorenal syndrome type 4 (CRS4) is a complication of chronic kidney disease. Panax notoginseng saponins (PNS) have been confirmed to be efficient in cardiovascular diseases. Our study aimed to explore the therapeutic role and mechanism of PNS in CRS4. METHODS CRS4 model rats and hypoxia-induced cardiomyocytes were treated with PNS, with and without pyroptosis inhibitor VX765 and ANRIL overexpression plasmids. Cardiac function and cardiorenal function biomarkers levels were measured by echocardiography and ELISA, respectively. Cardiac fibrosis was detected by Masson staining. Cell viability was determined by cell counting kit-8 and flow cytometry. Expression of fibrosis-related genes (COL-I, COL-III, TGF-β, α-SMA) and ANRIL was examined using RT-qPCR. Pyroptosis-related protein levels of NLRP3, ASC, IL-1β, TGF-β1, GSDMD-N, and caspase-1 were measured by western blotting or immunofluorescence staining. RESULTS PNS improved cardiac function, and inhibited cardiac fibrosis and pyroptosis in a dose-dependent manner in model rats and injured H9c2 cells (p < 0.01). The expression of fibrosis-related genes (COL-I, COL-III, TGF-β, α-SMA) and pyroptosis-related proteins (NLRP3, ASC, IL-1β, TGF-β1, GSDMD-N, and caspase-1) was inhibited by PNS in injured cardiac tissues and cells (p < 0.01). Additionally, ANRIL was upregulated in model rats and injured cells, but PNS reduced its expression in a dose-dependent manner (p < 0.05). Additionally, the inhibitory effect of PNS on pyroptosis in injured H9c2 cells was enhanced by VX765 and reversed by ANRIL overexpression, respectively (p < 0.05). CONCLUSION PNS inhibits pyroptosis by downregulating lncRNA-ANRIL in CRS4.
Collapse
Affiliation(s)
- Ying Xu
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Luxi Cao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Wenli Zou
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Rizhen Yu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Wei Shen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
126
|
Tanase DM, Valasciuc E, Gosav EM, Ouatu A, Buliga-Finis ON, Floria M, Maranduca MA, Serban IL. Portrayal of NLRP3 Inflammasome in Atherosclerosis: Current Knowledge and Therapeutic Targets. Int J Mol Sci 2023; 24:ijms24098162. [PMID: 37175869 PMCID: PMC10179095 DOI: 10.3390/ijms24098162] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
We are witnessing the globalization of a specific type of arteriosclerosis with rising prevalence, incidence and an overall cardiovascular disease burden. Currently, atherosclerosis increasingly affects the younger generation as compared to previous decades. While early preventive medicine has seen improvements, research advances in laboratory and clinical investigation promise to provide us with novel diagnosis tools. Given the physio-pathological complexity and epigenetic patterns of atherosclerosis and the discovery of new molecules involved, the therapeutic field of atherosclerosis has room for substantial growth. Thus, the scientific community is currently investigating the role of nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a crucial component of the innate immune system in different inflammatory disorders. NLRP3 is activated by distinct factors and numerous cellular and molecular events which trigger NLRP3 inflammasome assembly with subsequent cleavage of pro-interleukin (IL)-1β and pro-IL-18 pathways via caspase-1 activation, eliciting endothelial dysfunction, promotion of oxidative stress and the inflammation process of atherosclerosis. In this review, we introduce the basic cellular and molecular mechanisms of NLRP3 inflammasome activation and its role in atherosclerosis. We also emphasize its promising therapeutic pharmaceutical potential.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Oana Nicoleta Buliga-Finis
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Minela Aida Maranduca
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Department of Morpho-Functional Sciences II, Discipline of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
127
|
Huang J, Zhao J, Wang X, Ma L, Ma Z, Meng X, Fan H. SnRK1 signaling regulates cucumber growth and resistance to Corynespora cassiicola. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2023; 332:111716. [PMID: 37086974 DOI: 10.1016/j.plantsci.2023.111716] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Energy metabolism is one of the key factors determining the growth and development of plants and the response to biotic and abiotic stresses. Sucrose non-fermentation 1 related protein kinase 1 (SnRK1) is an important energy-sensitive regulator that plays a key role in the overall control of carbohydrate metabolism. However, little is known about the function of SnRK1 in cucumber. In this study, metformin (an SnRK1 activator) and trehalose (an SnRK1 inhibitor) were used to investigate the role of SnRK1 signaling in cucumber. The results showed that SnRK1 activation could inhibit the growth of cucumber, slow down the net photosynthetic rate (Pn), reduce the contents of photosynthetic pigments and soluble sugars, and suppress the expression of genes related to sucrose metabolism. By contrast, SnRK1 inhibition yielded opposite results. Furthermore, SnRK1 activation and CsSnRK1 over-expression improved cucumber resistance to Corynespora cassiicola. While, SnRK1 inhibition and CsSnRK1 silencing reduced the resistance of cucumber to C. cassiicola. The results indicated that CsSnRK1 gene can positively regulate the resistance of cucumber to C. cassiicola. We conclude that CsSnRK1 signaling plays an important role in balancing the growth and immune response of cucumber. These results can be applied to the improvement of disease-resistant cucumber varieties.
Collapse
Affiliation(s)
- Jingnan Huang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Juyong Zhao
- Liaoning Academy of Agricultural Sciences, Shenyang, Liaoning 110161, China
| | - Xue Wang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Lifeng Ma
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Zhangtong Ma
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Xiangnan Meng
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China; Key Laboratory of Protected Horticulture of Ministry of Education, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Haiyan Fan
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China; Key Laboratory of Protected Horticulture of Ministry of Education, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| |
Collapse
|
128
|
Zhang WJ, Li KY, Lan Y, Zeng HY, Chen SQ, Wang H. NLRP3 Inflammasome: A key contributor to the inflammation formation. Food Chem Toxicol 2023; 174:113683. [PMID: 36809826 DOI: 10.1016/j.fct.2023.113683] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/04/2023] [Accepted: 02/17/2023] [Indexed: 02/21/2023]
Abstract
Inflammation is an important part of the development of various organ diseases. The inflammasome, as an innate immune receptor, plays an important role in the formation of inflammation. Among various inflammasomes, the NLRP3 inflammasome is the most well studied. The NLRP3 inflammasome is composed of skeletal protein NLRP3, apoptosis-associated speck-like protein (ASC) and pro-caspase-1. There are three types of activation pathways: (1) "classical" activation pathway; (2) "non-canonical" activation pathway; (3) "alternative" activation pathway. The activation of NLRP3 inflammasome is involved in many inflammatory diseases. A variety of factors (such as genetic factors, environmental factors, chemical factors, viral infection, etc.) have been proved to activate NLRP3 inflammasome and promote the inflammatory response of the lung, heart, liver, kidney and other organs in the body. Especially, the mechanism of NLRP3 inflammation and its related molecules in its associated diseases remains not to be summarized, namely they may promote or delay inflammatory diseases in different cells and tissues. This article reviews the structure and function of the NLRP3 inflammasome and its role in various inflammations, including inflammations caused by chemically toxic substances.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Ke-Yun Li
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Yi Lan
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Han-Yi Zeng
- Department of Genetics, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Shui-Qin Chen
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, PR China.
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, PR China.
| |
Collapse
|
129
|
Sehrawat A, Mishra J, Mastana SS, Navik U, Bhatti GK, Reddy PH, Bhatti JS. Dysregulated autophagy: A key player in the pathophysiology of type 2 diabetes and its complications. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166666. [PMID: 36791919 DOI: 10.1016/j.bbadis.2023.166666] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
Autophagy is essential in regulating the turnover of macromolecules via removing damaged organelles, misfolded proteins in various tissues, including liver, skeletal muscles, and adipose tissue to maintain the cellular homeostasis. In these tissues, a specific type of autophagy maintains the accumulation of lipid droplets which is directly related to obesity and the development of insulin resistance. It appears to play a protective role in a normal physiological environment by eliminating the invading pathogens, protein aggregates, and damaged organelles and generating energy and new building blocks by recycling the cellular components. Ageing is also a crucial modulator of autophagy process. During stress conditions involving nutrient deficiency, lipids excess, hypoxia etc., autophagy serves as a pro-survival mechanism by recycling the free amino acids to maintain the synthesis of proteins. The dysregulated autophagy has been found in several ageing associated diseases including type 2 diabetes (T2DM), cancer, and neurodegenerative disorders. So, targeting autophagy can be a promising therapeutic strategy against the progression to diabetes related complications. Our article provides a comprehensive outline of understanding of the autophagy process, including its types, mechanisms, regulation, and role in the pathophysiology of T2DM and related complications. We also explored the significance of autophagy in the homeostasis of β-cells, insulin resistance (IR), clearance of protein aggregates such as islet amyloid polypeptide, and various insulin-sensitive tissues. This will further pave the way for developing novel therapeutic strategies for diabetes-related complications.
Collapse
Affiliation(s)
- Abhishek Sehrawat
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Jayapriya Mishra
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Umashanker Navik
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India.
| |
Collapse
|
130
|
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, Yang G, Zhang Z. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med 2023; 10:1088575. [PMID: 37063954 PMCID: PMC10090687 DOI: 10.3389/fcvm.2023.1088575] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic proteins and organelles, which realizes the metabolic needs of cells and the renewal of organelles. Autophagy-related genes (ATGs) are the main molecular mechanisms controlling autophagy, and their functions can coordinate the whole autophagic process. Autophagy can also play a role in cardiovascular disease through several key signaling pathways, including PI3K/Akt/mTOR, IGF/EGF, AMPK/mTOR, MAPKs, p53, Nrf2/p62, Wnt/β-catenin and NF-κB pathways. In this paper, we reviewed the signaling pathway of cross-interference between autophagy and cardiovascular diseases, and analyzed the development status of novel cardiovascular disease treatment by targeting the core molecular mechanism of autophagy as well as the critical signaling pathway. Induction or inhibition of autophagy through molecular mechanisms and signaling pathways can provide therapeutic benefits for patients. Meanwhile, we hope to provide a unique insight into cardiovascular treatment strategies by understanding the molecular mechanism and signaling pathway of crosstalk between autophagy and cardiovascular diseases.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuan Zhou
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Yang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linlin Wang
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Longfei Feng
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jin Xu
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Wang
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Haixiang Su
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - GuoWei Yang
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
131
|
AlTamimi JZ, AlFaris NA, Alshammari GM, Alagal RI, Aljabryn DH, Yahya MA. The Protective Effect of 11-Keto-β-Boswellic Acid against Diabetic Cardiomyopathy in Rats Entails Activation of AMPK. Nutrients 2023; 15:nu15071660. [PMID: 37049501 PMCID: PMC10097356 DOI: 10.3390/nu15071660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/25/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
This study examined the protective effect of 11-keto-β-boswellic acid (AKBA) against streptozotocin (STZ)-induced diabetic cardiomyopathy (DC) in rats and examined the possible mechanisms of action. Male rats were divided into 5 groups (n = 8/each): (1) control, AKBA (10 mg/kg, orally), STZ (65 mg/kg, i.p.), STZ + AKBA (10 mg/kg, orally), and STZ + AKBA + compound C (CC/an AMPK inhibitor, 0.2 mg/kg, i.p.). AKBA improved the structure and the systolic and diastolic functions of the left ventricles (LVs) of STZ rats. It also attenuated the increase in plasma glucose, plasma insulin, and serum and hepatic levels of triglycerides (TGs), cholesterol (CHOL), and free fatty acids (FFAs) in these diabetic rats. AKBA stimulated the ventricular activities of phosphofructokinase (PFK), pyruvate dehydrogenase (PDH), and acetyl CoA carboxylase (ACC); increased levels of malonyl CoA; and reduced levels of carnitine palmitoyltransferase I (CPT1), indicating improvement in glucose and FA oxidation. It also reduced levels of malondialdehyde (MDA); increased mitochondria efficiency and ATP production; stimulated mRNA, total, and nuclear levels of Nrf2; increased levels of glutathione (GSH), heme oxygenase (HO-1), superoxide dismutase (SOD), and catalase (CAT); but reduced the expression and nuclear translocation of NF-κB and levels of tumor-necrosis factor-α (TNF-α) and interleukin-6 (IL-6). These effects were concomitant with increased activities of AMPK in the LVs of the control and STZ-diabetic rats. Treatment with CC abolished all these protective effects of AKBA. In conclusion, AKBA protects against DC in rats, mainly by activating the AMPK-dependent control of insulin release, cardiac metabolism, and antioxidant and anti-inflammatory effects.
Collapse
|
132
|
Ke D, Zhang Z, Liu J, Chen P, Li J, Sun X, Chu Y, Li L. Ferroptosis, necroptosis and cuproptosis: Novel forms of regulated cell death in diabetic cardiomyopathy. Front Cardiovasc Med 2023; 10:1135723. [PMID: 36970345 DOI: 10.3389/fcvm.2023.1135723if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 02/22/2023] [Indexed: 09/15/2024] Open
Abstract
Diabetes is a common chronic metabolic disease, and its incidence continues to increase year after year. Diabetic patients mainly die from various complications, with the most common being diabetic cardiomyopathy. However, the detection rate of diabetic cardiomyopathy is low in clinical practice, and targeted treatment is lacking. Recently, a large number of studies have confirmed that myocardial cell death in diabetic cardiomyopathy involves pyroptosis, apoptosis, necrosis, ferroptosis, necroptosis, cuproptosis, cellular burial, and other processes. Most importantly, numerous animal studies have shown that the onset and progression of diabetic cardiomyopathy can be mitigated by inhibiting these regulatory cell death processes, such as by utilizing inhibitors, chelators, or genetic manipulation. Therefore, we review the role of ferroptosis, necroptosis, and cuproptosis, three novel forms of cell death in diabetic cardiomyopathy, searching for possible targets, and analyzing the corresponding therapeutic approaches to these targets.
Collapse
Affiliation(s)
- Dan Ke
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
- School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, China
| | - Jieting Liu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Peijian Chen
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Jialing Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Xinhai Sun
- Department of Thoracic Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
133
|
Ke D, Zhang Z, Liu J, Chen P, Li J, Sun X, Chu Y, Li L. Ferroptosis, necroptosis and cuproptosis: Novel forms of regulated cell death in diabetic cardiomyopathy. Front Cardiovasc Med 2023; 10:1135723. [PMID: 36970345 PMCID: PMC10036800 DOI: 10.3389/fcvm.2023.1135723] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Diabetes is a common chronic metabolic disease, and its incidence continues to increase year after year. Diabetic patients mainly die from various complications, with the most common being diabetic cardiomyopathy. However, the detection rate of diabetic cardiomyopathy is low in clinical practice, and targeted treatment is lacking. Recently, a large number of studies have confirmed that myocardial cell death in diabetic cardiomyopathy involves pyroptosis, apoptosis, necrosis, ferroptosis, necroptosis, cuproptosis, cellular burial, and other processes. Most importantly, numerous animal studies have shown that the onset and progression of diabetic cardiomyopathy can be mitigated by inhibiting these regulatory cell death processes, such as by utilizing inhibitors, chelators, or genetic manipulation. Therefore, we review the role of ferroptosis, necroptosis, and cuproptosis, three novel forms of cell death in diabetic cardiomyopathy, searching for possible targets, and analyzing the corresponding therapeutic approaches to these targets.
Collapse
Affiliation(s)
- Dan Ke
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
- School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, China
| | - Jieting Liu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Peijian Chen
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Jialing Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Xinhai Sun
- Department of Thoracic Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
- Correspondence: Yanhui Chu Luxin Li
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
- Correspondence: Yanhui Chu Luxin Li
| |
Collapse
|
134
|
Seksaria S, Mehan S, Dutta BJ, Gupta GD, Ganti SS, Singh A. Oxymatrine and insulin resistance: Focusing on mechanistic intricacies involve in diabetes associated cardiomyopathy via SIRT1/AMPK and TGF-β signaling pathway. J Biochem Mol Toxicol 2023; 37:e23330. [PMID: 36890713 DOI: 10.1002/jbt.23330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/03/2023] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
Cardiomyopathy (CDM) and related morbidity and mortality are increasing at an alarming rate, in large part because of the increase in the number of diabetes mellitus cases. The clinical consequence associated with CDM is heart failure (HF) and is considerably worse for patients with diabetes mellitus, as compared to nondiabetics. Diabetic cardiomyopathy (DCM) is characterized by structural and functional malfunctioning of the heart, which includes diastolic dysfunction followed by systolic dysfunction, myocyte hypertrophy, cardiac dysfunctional remodeling, and myocardial fibrosis. Indeed, many reports in the literature indicate that various signaling pathways, such as the AMP-activated protein kinase (AMPK), silent information regulator 1 (SIRT1), PI3K/Akt, and TGF-β/smad pathways, are involved in diabetes-related cardiomyopathy, which increases the risk of functional and structural abnormalities of the heart. Therefore, targeting these pathways augments the prevention as well as treatment of patients with DCM. Alternative pharmacotherapy, such as that using natural compounds, has been shown to have promising therapeutic effects. Thus, this article reviews the potential role of the quinazoline alkaloid, oxymatrine obtained from the Sophora flavescensin CDM associated with diabetes mellitus. Numerous studies have given a therapeutic glimpse of the role of oxymatrine in the multiple secondary complications related to diabetes, such as retinopathy, nephropathy, stroke, and cardiovascular complications via reductions in oxidative stress, inflammation, and metabolic dysregulation, which might be due to targeting signaling pathways, such as AMPK, SIRT1, PI3K/Akt, and TGF-β pathways. Thus, these pathways are considered central regulators of diabetes and its secondary complications, and targeting these pathways with oxymatrine might provide a therapeutic tool for the diagnosis and treatment of diabetes-associated cardiomyopathy.
Collapse
Affiliation(s)
- Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Bhaskar J Dutta
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Ghanshyam D Gupta
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Subrahmanya S Ganti
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab, India
| |
Collapse
|
135
|
Deng J, Yan F, Tian J, Qiao A, Yan D. Potential clinical biomarkers and perspectives in diabetic cardiomyopathy. Diabetol Metab Syndr 2023; 15:35. [PMID: 36871006 PMCID: PMC9985231 DOI: 10.1186/s13098-023-00998-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a serious cardiovascular complication and the leading cause of death in diabetic patients. Patients typically do not experience any symptoms and have normal systolic and diastolic cardiac functions in the early stages of DCM. Because the majority of cardiac tissue has already been destroyed by the time DCM is detected, research must be conducted on biomarkers for early DCM, early diagnosis of DCM patients, and early symptomatic management to minimize mortality rates among DCM patients. Most of the existing implemented clinical markers are not very specific for DCM, especially in the early stages of DCM. Recent studies have shown that a number of new novel markers, such as galactin-3 (Gal-3), adiponectin (APN), and irisin, have significant changes in the clinical course of the various stages of DCM, suggesting that we may have a positive effect on the identification of DCM. As a summary of the current state of knowledge regarding DCM biomarkers, this review aims to inspire new ideas for identifying clinical markers and related pathophysiologic mechanisms that could be used in the early diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Jianxin Deng
- Department of Endocrinology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen Clinical Research Center for Metabolic Diseases, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, Guangdong Province, China
| | - Fang Yan
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, 611137, Sichuan Province, China
| | - Jinglun Tian
- Department of Geriatrics, the Traditional Chinese Medicine Hospital of Wenjiang District, Chengdu, 611130, China
| | - Aijun Qiao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, Guangdong Province, China.
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen Clinical Research Center for Metabolic Diseases, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, Guangdong Province, China.
| |
Collapse
|
136
|
Zhang L, Cui T, Wang X. The Interplay Between Autophagy and Regulated Necrosis. Antioxid Redox Signal 2023; 38:550-580. [PMID: 36053716 PMCID: PMC10025850 DOI: 10.1089/ars.2022.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022]
Abstract
Significance: Autophagy is critical to cellular homeostasis. Emergence of the concept of regulated necrosis, such as necroptosis, ferroptosis, pyroptosis, and mitochondrial membrane-permeability transition (MPT)-derived necrosis, has revolutionized the research into necrosis. Both altered autophagy and regulated necrosis contribute to major human diseases. Recent studies reveal an intricate interplay between autophagy and regulated necrosis. Understanding the interplay at the molecular level will provide new insights into the pathophysiology of related diseases. Recent Advances: Among the three forms of autophagy, macroautophagy is better studied for its crosstalk with regulated necrosis. Macroautophagy seemingly can either antagonize or promote regulated necrosis, depending upon the form of regulated necrosis, the type of cells or stimuli, and other cellular contexts. This review will critically analyze recent advances in the molecular mechanisms governing the intricate dialogues between macroautophagy and main forms of regulated necrosis. Critical Issues: The dual roles of autophagy, either pro-survival or pro-death characteristics, intricate the mechanistic relationship between autophagy and regulated necrosis at molecular level in various pathological conditions. Meanwhile, key components of regulated necrosis are also involved in the regulation of autophagy, which further complicates the interrelationship. Future Directions: Resolving the controversies over causation between altered autophagy and a specific form of regulated necrosis requires approaches that are more definitive, where rigorous evaluation of autophagic flux and the development of more reliable and specific methods to quantify each form of necrosis will be essential. The relationship between chaperone-mediated autophagy or microautophagy and regulated necrosis remains largely unstudied. Antioxid. Redox Signal. 38, 550-580.
Collapse
Affiliation(s)
- Lei Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, The University of South Dakota Sanford School of Medicine, Vermillion, South Dakota, USA
| |
Collapse
|
137
|
Sodium butyrate attenuate hyperglycemia-induced inflammatory response and renal injury in diabetic mice. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:121-132. [PMID: 36692460 DOI: 10.2478/acph-2023-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 01/25/2023]
Abstract
The activation of the monocyte-macrophage system and the damage to the renal and pancreatic tissue are common complications in patients with diabetes induced by hyper-glycemia. This study aimed to evaluate the effect and mechanism of butyrate (NaB), a metabolite of intestinal flora, on inhibiting the inflammatory response of human monocyte-macrophages (THP-1 cells) induced by high glucose and the damage of pancreatic and renal tissue in diabetic mice. The results showed that high concentration glucose significantly up-regulated the expressions of IL-1β, TNF-α, and NLRP3 in THP-1 cells and mouse spleen, and that NaB could inhibit the overexpression of those genes. The abundance of Beclin-1, LC3B and reactive oxygen species (ROS) in THP-1 cells is increased due to the high glucose concentration, and NaB can inhibit the genes responsible for upregulating the expression. In diabetic mice, vacuolar degeneration of renal tubules was observed. Then we observed that some of the epithelial cells of the renal tubules were exfoliated and some formed tubules. NaB could alleviate these pathological lesions, but NaB cannot alleviate pancreatic injury. Our results indicated that NaB could be used for the prevention and adjuvant treatment of diabetic kidney injury.
Collapse
|
138
|
Peng C, Zhang Y, Lang X, Zhang Y. Role of mitochondrial metabolic disorder and immune infiltration in diabetic cardiomyopathy: new insights from bioinformatics analysis. J Transl Med 2023; 21:66. [PMID: 36726122 PMCID: PMC9893675 DOI: 10.1186/s12967-023-03928-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is one of the common cardiovascular complications of diabetes and a leading cause of death in diabetic patients. Mitochondrial metabolism and immune-inflammation are key for DCM pathogenesis, but their crosstalk in DCM remains an open issue. This study explored the separate roles of mitochondrial metabolism and immune microenvironment and their crosstalk in DCM with bioinformatics. METHODS DCM chip data (GSE4745, GSE5606, and GSE6880) were obtained from NCBI GEO, while mitochondrial gene data were downloaded from MitoCarta3.0 database. Differentially expressed genes (DEGs) were screened by GEO2R and processed for GSEA, GO and KEGG pathway analyses. Mitochondria-related DEGs (MitoDEGs) were obtained. A PPI network was constructed, and the hub MitoDEGs closely linked to DCM or heart failure were identified with CytoHubba, MCODE and CTD scores. Transcription factors and target miRNAs of the hub MitoDEGs were predicted with Cytoscape and miRWalk database, respectively, and a regulatory network was established. The immune infiltration pattern in DCM was analyzed with ImmuCellAI, while the relationship between MitoDEGs and immune infiltration abundance was investigated using Spearman method. A rat model of DCM was established to validate the expression of hub MitoDEGs and their relationship with cardiac function. RESULTS MitoDEGs in DCM were significantly enriched in pathways involved in mitochondrial metabolism, immunoregulation, and collagen synthesis. Nine hub MitoDEGs closely linked to DCM or heart failure were obtained. Immune analysis revealed significantly increased infiltration of B cells while decreased infiltration of DCs in immune microenvironment of DCM. Spearman analysis demonstrated that the hub MitoDEGs were positively associated with the infiltration of pro-inflammatory immune cells, but negatively associated with the infiltration of anti-inflammatory or regulatory immune cells. In the animal experiment, 4 hub MitoDEGs (Pdk4, Hmgcs2, Decr1, and Ivd) showed an expression trend consistent with bioinformatics analysis result. Additionally, the up-regulation of Pdk4, Hmgcs2, Decr1 and the down-regulation of Ivd were distinctly linked to reduced cardiac function. CONCLUSIONS This study unraveled the interaction between mitochondrial metabolism and immune microenvironment in DCM, providing new insights into the research on potential pathogenesis of DCM and the exploration of novel targets for medical interventions.
Collapse
Affiliation(s)
- Cheng Peng
- grid.412463.60000 0004 1762 6325Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150001 China ,grid.410736.70000 0001 2204 9268Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001 China
| | - Yanxiu Zhang
- grid.412463.60000 0004 1762 6325Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150001 China ,grid.410736.70000 0001 2204 9268Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001 China
| | - Xueyan Lang
- grid.412463.60000 0004 1762 6325Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150001 China ,grid.410736.70000 0001 2204 9268Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001 China
| | - Yao Zhang
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China. .,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
139
|
Klotho improves cardiac fibrosis, inflammatory cytokines, ferroptosis, and oxidative stress in mice with myocardial infarction. J Physiol Biochem 2023:10.1007/s13105-023-00945-5. [PMID: 36701072 DOI: 10.1007/s13105-023-00945-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023]
Abstract
The anti-aging protein Klotho has been associated with cardiovascular health protection. Nevertheless, the protective mechanism remains unknown. The present study is aimed at exploring the effect of Klotho on cardiac remodeling and its potential mechanism in mice with myocardial infarction (MI). We used left anterior coronary artery descending ligation to develop an MI model for in vivo analyses. In contrast, H9C2 cells and cardiac fibroblasts were used to establish the oxygen-glucose deprivation (OGD) model in in vitro analyses. In vivo and in vitro models were treated with Klotho. Compound C, an AMPK signaling inhibitor, was used to determine whether Klotho's effects are mediated through the AMPK/mTOR signaling pathway. Echocardiography, Masson trichrome staining, immunofluorescence, immunohistochemistry, real-time polymerase chain reaction (RT-PCR), and western blot were used to detect the related indicators. The findings of the in vivo model indicate that Klotho treatment improved the mice's cardiac function, reduced cardiac fibrosis, and attenuated myocardial inflammatory factors, ferroptosis, and oxidative stress. The results of the in vitro model were in line with the findings of in vivo modeling. An AMPK inhibitor, Compound C, reversed all these effects. In conclusion, Klotho potentially improves cardiac remodeling in MI mice by regulating AMPK/mTOR signaling, demonstrating Klotho as an effective MI therapeutic agent.
Collapse
|
140
|
Abstract
BACKGROUND Metformin has good anti-hyperglycemic effectiveness, but does not induce hypoglycemia,is very safe, and has become the preferred drug for the treatment of type 2 diabetes. Recently, the other effects of metformin, such as being anti-inflammatory and delaying aging, have also attracted increased attention. METHODS AND RESULTS The relevant literatures on pubmed and other websites for reading, classification and sorting, and did not involve any animal experiments. CONCLUSION Metformin has anti-inflammatory effects through multiple routes, which provides potential therapeutic targets for certain inflammatory diseases, such as neuroinflammation and rheumatoid arthritis. In addition, inflammation is a key component of tumor occurrence and development ; thus, targeted inflammatory intervention is a significant benefit for both cancer prevention and treatment. Therefore, metformin may have further potential for inflammation-related disease prevention and treatmen. However, the inflammatory mechanism is complex; various molecules are connected and influence each other. For example, metformin significantly inhibits p65 nuclear translocation, but pretreatment with compound C, an AMPK inhibitor, abolishes this effect, and silencing of HMGB1 inhibits NF-κB activation . SIRT1 deacetylates FoxO, increasing its transcriptional activity . mTOR in dendritic cells regulates FoxO1 via AKT. The interactions among various molecules should be further explored to clarify their specific mechanisms and provide more direction for the treatment of inflammatory diseases, as well as cancer.
Collapse
|
141
|
Zhou J, Yan S, Guo X, Gao Y, Chen S, Li X, Zhang Y, Wang Q, Zheng T, Chen L. Salidroside protects pancreatic β-cells against pyroptosis by regulating the NLRP3/GSDMD pathway in diabetic conditions. Int Immunopharmacol 2023; 114:109543. [PMID: 36508922 DOI: 10.1016/j.intimp.2022.109543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
The NACHT, LRP, and PYD domains-containing protein 3 (NLRP3) inflammasome-evoked chronic inflammation is involved in the pathogenesis of diabetes mellitus (DM), and the NLRP3/gasdermin D (GSDMD)-mediated canonical pathway of pyroptosis leads to the loss of pancreatic β-cells and failure of pancreatic function in DM. A previous study demonstrated that salidroside (SAL) alleviates the pathological hyperplasia of pancreatic β-cells in db/db mice. However, it is not clear whether the NLRP3/GSDMD pathway-mediated pyroptosis can be regulated by SAL. In addition, the action of SAL on pancreatic β-cells in DM remains poorly understood. Thus, this study aimed to investigate the effects and underlying mechanisms of SAL on pancreatic β-cell pyroptosis. Rat insulinoma (INS-1) cells were cultured in a medium containing either high glucose (HG) or HG plus high insulin (HG-HI), and the effects of SAL on cell viability, AMP-activated protein kinase (AMPK) activity, reactive oxygen species (ROS) generation, NLRP3/GSDMD activation, and pyroptotic body formation were assessed. Streptozocin-induced DM mice were used to further investigate the effects of SAL on pancreatic pyroptosis. The results revealed aberrances on cell viability, AMPK activity, ROS generation, NLRP3/GSDMD activation, and pyroptotic body formation in HG- and HG-HI-exposed INS-1 cells; these abnormal effects were corrected by SAL in both a concentration- and AMPK-dependent manner. Moreover, SAL administration activated AMPK, suppressed NLRP3/GSDMD signaling, and protected pancreatic β-cells against pyroptosis in DM mice. These findings suggest that SAL promotes AMPK activation to suppress NLRP3/GSDMD-related pyroptosis in pancreatic β-cells under DM conditions.
Collapse
Affiliation(s)
- Jun Zhou
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shan Yan
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xu Guo
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yanguo Gao
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shiqi Chen
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaohan Li
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yonghong Zhang
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qibin Wang
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Tao Zheng
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Li Chen
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
142
|
Liu C, Liu Y, Chen H, Yang X, Lu C, Wang L, Lu J. Myocardial injury: where inflammation and autophagy meet. BURNS & TRAUMA 2023; 11:tkac062. [PMID: 36873283 PMCID: PMC9977361 DOI: 10.1093/burnst/tkac062] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/23/2022] [Indexed: 03/04/2023]
Abstract
Autophagy is a highly conserved bulk degradation mechanism that degrades damaged organelles, aged proteins and intracellular contents to maintain the homeostasis of the intracellular microenvironment. Activation of autophagy can be observed during myocardial injury, during which inflammatory responses are strongly triggered. Autophagy can inhibit the inflammatory response and regulate the inflammatory microenvironment by removing invading pathogens and damaged mitochondria. In addition, autophagy may enhance the clearance of apoptotic and necrotic cells to promote the repair of damaged tissue. In this paper, we briefly review the role of autophagy in different cell types in the inflammatory microenvironment of myocardial injury and discuss the molecular mechanism of autophagy in regulating the inflammatory response in a series of myocardial injury conditions, including myocardial ischemia, ischemia/reperfusion injury and sepsis cardiomyopathy.
Collapse
Affiliation(s)
- Chunping Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 51080, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, 510080, China
| | - Yanjiao Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 51080, China
| | - Huiqi Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 51080, China
| | - Xiaofei Yang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 51080, China
| | - Chuanjian Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 51080, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, 510080, China
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 51080, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
143
|
Chen J, Zhu G, Xiao W, Huang X, Wang K, Zong Y. Ginsenoside Rg1 Ameliorates Pancreatic Injuries via the AMPK/mTOR Pathway in vivo and in vitro. Diabetes Metab Syndr Obes 2023; 16:779-794. [PMID: 36945297 PMCID: PMC10024876 DOI: 10.2147/dmso.s401642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND The main propanaxatriol-type saponin found in ginseng (Panax ginseng C. A. Mey), ginsenoside Rg1 (G-Rg1), has bioactivities that include anti-inflammatory, antioxidant, and anti-diabetic properties. This study aimed to investigate the effects of G-Rg1 on streptozotocin (STZ)-induced Type 1 Diabetes mellitus (T1DM) mice and the insulin-secreting cell line in RIN-m5F cells with high-glucose (HG) treatment. METHODS The STZ-induced DM mice model was treated with G-Rg1 alone or combined with 3-Methyladenine (3-MA, an autophagy inhibitor)/rapamycin (RAPA, an autophagy activator) for 8 weeks, and levels of glucose and lipid metabolism, histopathological changes, as well as autophagy and apoptosis of relevant markers were estimated. In vitro, the HG-induced RIN-m5F cells were treated with G-Rg1, 3-MA, and Compound C (CC), an AMPK inhibitor, or their combinations to estimate the influences on cell apoptosis, autophagy, and AMPK/mTOR pathway-associated target gene levels. RESULTS G-Rg1 treatment attenuated glucose and lipid metabolism disorder and pancreatic fibrosis in diabetic mice. In addition, subdued autophagy and p-AMPK protein expression, and enhanced p-mTOR protein expression and apoptosis levels in TIDM mice and HG-induced RIN-m5F cells were ameliorated by G-Rg1 treatment. Furthermore, these anti-apoptosis effects of G-Rg1 were partially abolished by 3-MA and CC. CONCLUSION Our findings revealed that G-Rg1 exhibits strong anti-apoptosis ability in pancreatic tissues of type 1 diabetic mice and HG-induced RIN-m5F cells, and the mechanisms involved in activating AMPK and inhibiting mTOR-mediated autophagy, indicating that G-Rg1 may have the therapeutic and preventive potential for treating pancreatic injury in diabetic patients.
Collapse
Affiliation(s)
- Jin Chen
- Department of Hematology, Yiwu Central Hospital, Yiwu, People’s Republic of China
| | - Guoping Zhu
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
| | - Wenbo Xiao
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xiaosong Huang
- Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
| | - Kewu Wang
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
- Correspondence: Kewu Wang; Yi Zong, Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, No. N1, Shangcheng Avenue, Yiwu, Zhejiang, People’s Republic of China, Email ;
| | - Yi Zong
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People’s Republic of China
| |
Collapse
|
144
|
Wang Q, Chen T, Shuqing Z, Yu L, Chen S, Lu H, Zhu H, Min X, Li X, Liu L. Xanthohumol relieves arthritis pain in mice by suppressing mitochondrial-mediated inflammation. Mol Pain 2023; 19:17448069231204051. [PMID: 37699859 PMCID: PMC10536840 DOI: 10.1177/17448069231204051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/19/2023] [Accepted: 09/11/2023] [Indexed: 09/14/2023] Open
Abstract
Chronic pain is the most common symptom for people who suffer from rheumatoid arthritis and it affects approximately 1% of the global population. Neuroinflammation in the spinal cord induces chronic arthritis pain. In this study, a collagen-induced arthritis (CIA) mice model was established through intradermally injection of type II collagen in complete Freund's adjuvant solution. Following CIA inducement, the paws and ankles of mice were found to swell, mechanical pain and spontaneous pain were induced, and their motor coordination was impaired. The spinal inflammatory reaction was triggered, which presented as severe infiltration of inflammatory cells, and the expression levels of GFAP, IL-1β, NLRP3, and cleaved caspase-1 increased. Oxidative stress in the spinal cord of CIA mice was manifested as reduced Nrf2 and NDUFB11 expression and SOD activity, and increased levels of DHODH and Cyto-C. At the same time, spinal AMPK activity was decreased. In order to explore the potential therapeutic options for arthritic pain, Xanthohumol (Xn) was intraperitoneally injected into mice for three consecutive days. Xn treatment was found to reduce the number of spontaneous flinches, in addition to elevating mechanical pain thresholds and increasing latency time. At the same time, Xn treatment in the spinal cord reduced NLRP3 inflammasome-mediated inflammation, increased the Nrf2-mediated antioxidant response, and decreased mitochondrial ROS level. In addition, Xn was found to bind with AMPK via two electrovalent bonds and increased AMPK phosphorylation at Thr174. In summary, the findings indicate that Xn treatment activates AMPK, increases Nrf2-mediated antioxidant response, reduces Drp1-mediated mitochondrial dysfunction, suppresses neuroinflammation, and can serve to relieve arthritis pain.
Collapse
Affiliation(s)
- Qin Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Tao Chen
- Xianning Central Hospital, First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Zhen Shuqing
- Matang Hospital of Traditional Chinese Medicine, Xianning, China
| | - Liangzhu Yu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Shaohui Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hong Lu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Haili Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xie Min
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xiong Li
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ling Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
145
|
Effects of Metformin Delivery via Biomaterials on Bone and Dental Tissue Engineering. Int J Mol Sci 2022; 23:ijms232415905. [PMID: 36555544 PMCID: PMC9779818 DOI: 10.3390/ijms232415905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022] Open
Abstract
Bone tissue engineering is a promising approach that uses seed-cell-scaffold drug delivery systems to reconstruct bone defects caused by trauma, tumors, or other diseases (e.g., periodontitis). Metformin, a widely used medication for type II diabetes, has the ability to enhance osteogenesis and angiogenesis by promoting cell migration and differentiation. Metformin promotes osteogenic differentiation, mineralization, and bone defect regeneration via activation of the AMP-activated kinase (AMPK) signaling pathway. Bone tissue engineering depends highly on vascular networks for adequate oxygen and nutrition supply. Metformin also enhances vascular differentiation via the AMPK/mechanistic target of the rapamycin kinase (mTOR)/NLR family pyrin domain containing the 3 (NLRP3) inflammasome signaling axis. This is the first review article on the effects of metformin on stem cells and bone tissue engineering. In this paper, we review the cutting-edge research on the effects of metformin on bone tissue engineering. This includes metformin delivery via tissue engineering scaffolds, metformin-induced enhancement of various types of stem cells, and metformin-induced promotion of osteogenesis, angiogenesis, and its regulatory pathways. In addition, the dental, craniofacial, and orthopedic applications of metformin in bone repair and regeneration are also discussed.
Collapse
|
146
|
Zhu N, Huang B, Zhu L. Bibliometric analysis of the inflammation in diabetic cardiomyopathy. Front Cardiovasc Med 2022; 9:1006213. [PMID: 36582738 PMCID: PMC9792483 DOI: 10.3389/fcvm.2022.1006213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Background Maladaptive inflammation is implicated in the development of diabetic cardiomyopathy (DCM). This study aimed to visually analyze the global scientific output over the past two decades regarding research on inflammation associated with DCM. Methods All relevant articles and reviews were retrieved in the Web of Science (WOS) Core Collection (limited to SCIE) using "inflammation" and "diabetic cardiomyopathy" as search terms. Articles and reviews published from 1 January 2001 to 28 February 2021 were collected. Visualization analysis and statistical analysis were conducted by Microsoft 365 Excel and VOSviewer 1.6.18. Results A total of 578 documents were finally selected for further analysis. The publications regarding inflammation and DCM increased gradually over approximately 20 years. The most prolific country was China, with 296 documents and the most citations (9,366). The most influential author groups were Lu Cai and Yihui Tan who were from the United States. The bibliometric analysis of co-occurrence keywords showed that inflammation in DCM is composed of numerous molecules (NF-κB, NLRP3 inflammasome, Nrf-2, TNF-α, protein kinase C, PPARα, TLR4, p38 mitogen-activated protein kinase, TGF-β, Sirt1, and AKT), a variety of cardiac cell types (stem cell, fibroblast, and cardiomyocyte), physiological processes (apoptosis, oxidative stress, autophagy, endoplasmic reticulum stress, hypertrophy, mitochondrion dysfunction, and proliferation), and drugs (sulforaphane, metformin, empagliflozin, and rosuvastatin). Conclusion Our bibliometric analysis presents the characteristics and trends of inflammation in DCM and shows that research on inflammation in DCM will continue to be a hotspot.
Collapse
Affiliation(s)
- Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, Wenzhou People’s Hospital, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China,*Correspondence: Ning Zhu,
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liuyan Zhu
- Department of General Practice, The Third Affiliated Hospital of Shanghai University, Wenzhou People’s Hospital, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
147
|
Du Y, Zhu YJ, Zhou YX, Ding J, Liu JY. Metformin in therapeutic applications in human diseases: its mechanism of action and clinical study. MOLECULAR BIOMEDICINE 2022; 3:41. [PMID: 36484892 PMCID: PMC9733765 DOI: 10.1186/s43556-022-00108-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Metformin, a biguanide drug, is the most commonly used first-line medication for type 2 diabetes mellites due to its outstanding glucose-lowering ability. After oral administration of 1 g, metformin peaked plasma concentration of approximately 20-30 μM in 3 h, and then it mainly accumulated in the gastrointestinal tract, liver and kidney. Substantial studies have indicated that metformin exerts its beneficial or deleterious effect by multiple mechanisms, apart from AMPK-dependent mechanism, also including several AMPK-independent mechanisms, such as restoring of redox balance, affecting mitochondrial function, modulating gut microbiome and regulating several other signals, such as FBP1, PP2A, FGF21, SIRT1 and mTOR. On the basis of these multiple mechanisms, researchers tried to repurpose this old drug and further explored the possible indications and adverse effects of metformin. Through investigating with clinical studies, researchers concluded that in addition to decreasing cardiovascular events and anti-obesity, metformin is also beneficial for neurodegenerative disease, polycystic ovary syndrome, aging, cancer and COVID-19, however, it also induces some adverse effects, such as gastrointestinal complaints, lactic acidosis, vitamin B12 deficiency, neurodegenerative disease and offspring impairment. Of note, the dose of metformin used in most studies is much higher than its clinically relevant dose, which may cast doubt on the actual effects of metformin on these disease in the clinic. This review summarizes these research developments on the mechanism of action and clinical evidence of metformin and discusses its therapeutic potential and clinical safety.
Collapse
Affiliation(s)
- Yang Du
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ya-Juan Zhu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yi-Xin Zhou
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jing Ding
- grid.54549.390000 0004 0369 4060Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan China
| | - Ji-Yan Liu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
148
|
Mo DG, Wang L, Han QF, Yu K, Liu JH, Yao HC. NLRP3 Inflammasome May Be a Biomarker for Risk Stratification in Patients with Acute Coronary Syndrome. J Inflamm Res 2022; 15:6595-6605. [PMID: 36510493 PMCID: PMC9739063 DOI: 10.2147/jir.s383903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose Acute coronary syndrome (ACS) has a high incidence and mortality rate worldwide, which has a considerable negative impact on the global economy. This study aimed to identify a group of ACS patients at a high risk of recurrent adverse cardiac events using the plasma NLRP3 inflammasome. Patients and methods ACS patients admitted to Liaocheng People's Hospital between June 2021 and March 2022 were enrolled in this study. Patients were divided into low (levels < 3.84 ng/mL) and high (levels ≥ 3.84 ng/mL) groups based on the median NLRP3 inflammasome levels. The patients were divided into three groups according to the Thrombolysis in Myocardial Infarction Risk Score for Secondary Prevention (TRS-2P): low (scores ≤ 2 points), intermediate (scores = 3 points), and high (score ≥ 4 points) risk. We investigated the relationship between NLRP3 inflammasome and laboratory indicators. Additionally, we examined whether the NLRP3 inflammasome was an independent predictor of high TRS-2P and explored the applicability of the plasma NLRP3 inflammasome for predicting high TRS-2P. Results Logistic regression analysis revealed that NLRP3 inflammasome was an independent predictor of high TRS-2P (odds ratio [OR]:2.013; 95% confidence interval [CI]: 1.174-3.452). The area under the receiver operating characteristic curve value of the NLRP3 inflammasome was 0.674 (95% CI: 0.611-0.737; P < 0.001). Conclusion NLRP3 inflammasome levels are an independent predictive factor for high TRS-2P levels, which indicates that the NLRP3 inflammasome may help predict the prognosis of ACS patients.
Collapse
Affiliation(s)
- De-Gang Mo
- Department of Cardiology, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, People’s Republic of China
| | - Lin Wang
- Cardiologic Color Doppler Room, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, People’s Republic of China
| | - Qian-Feng Han
- Department of Cardiology, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, People’s Republic of China
| | - Kang Yu
- Department of Laboratory Medicine, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, People’s Republic of China
| | - Jia-Hui Liu
- Department of Cardiology, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, People’s Republic of China
| | - Heng-Chen Yao
- Department of Cardiology, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, People’s Republic of China,Correspondence : Heng-Chen Yao, Department of Cardiology, Liaocheng People’s Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, People’s Republic of China, Email
| |
Collapse
|
149
|
Li JY, Zhao CC, Peng JF, Zhang M, Wang L, Yin G, Zhou P. The Protective Effect of Sheng Mai Yin on Diabetic Cardiomyopathy via NLRP3/Caspase-1 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:1234434. [PMID: 36506810 PMCID: PMC9731757 DOI: 10.1155/2022/1234434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/30/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022]
Abstract
Sheng Mai Yin (SMY) has therapeutic effects on myocardial infarction (MI), heart failure (HF), diabetic cardiomyopathy (DCM), and myocarditis. To study whether SMY can relieve pyroptosis and play a protective role in diabetic cardiomyopathy, a molecular docking technique was used to predict the possible mechanism of SMY against DCM. Then, a DCM rat model was induced by intraperitoneal injection of streptozotocin (STZ), divided into 5 groups: the DM group (model), SMY-L group (2.7 mL/kg SMY), SMY-M group (5.4 mL/kg SMY), SMY-H group (10.8 mL/kg SMY), and Met group (120 mg/kg metformin). Rats in the CTL group (control) and DM group were given normal saline. After 8 weeks, the levels of blood glucose, lipids, and myocardial enzymes were detected according to the kit instructions. Cardiac function was detected by echocardiography. HE and Masson were used to observing the pathological changes, collagen deposition, and collagen volume fraction (CVF). The apoptosis rate of cardiomyocytes was determined by Tunel. The IL-1β level was determined by ELISA and RT-PCR. The expressions of NLRP3, caspase-1, and GSDMD were measured using RT-PCR and Western blotting. The docking results suggested that SMY may act on NLRP3 and its downstream signal pathway. The in vivo results showed that SMY could reduce blood glucose and lipid levels, improve heart function, improve histopathological changes and myocardial enzymes, and alleviate cardiomyocyte apoptosis and myocardial fibrosis. SMY inhibited the mRNA and protein expressions of NLRP3, ASC, Caspase-1, and GSDMD and IL-1β production. SMY can reduce DCM by regulating the NLRP3/caspase-1 signaling pathway, providing a new research direction for the treatment of DCM.
Collapse
Affiliation(s)
- Jing-Ya Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Chun-Chun Zhao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jian-Fei Peng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Meng Zhang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Liang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Gang Yin
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
150
|
Exosomes derived from human umbilical cord mesenchymal stem cells (HUCMSC-EXO) regulate autophagy through AMPK-ULK1 signaling pathway to ameliorate diabetic cardiomyopathy. Biochem Biophys Res Commun 2022; 632:195-203. [PMID: 36240643 DOI: 10.1016/j.bbrc.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/19/2022]
|