1
|
Zhang C, Shi Y, Liu C, Sudesh SM, Hu Z, Li P, Liu Q, Ma Y, Shi A, Cai H. Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease. Cardiovasc Diabetol 2024; 23:169. [PMID: 38750502 PMCID: PMC11097480 DOI: 10.1186/s12933-024-02273-4] [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: 10/12/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
Diabetic heart disease (DHD) is a serious complication in patients with diabetes. Despite numerous studies on the pathogenic mechanisms and therapeutic targets of DHD, effective means of prevention and treatment are still lacking. The pathogenic mechanisms of DHD include cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Macrophages, the primary cells of the human innate immune system, contribute significantly to these pathological processes, playing an important role in human disease and health. Therefore, drugs targeting macrophages hold great promise for the treatment of DHD. In this review, we examine how macrophages contribute to the development of DHD and which drugs could potentially be used to target macrophages in the treatment of DHD.
Collapse
Affiliation(s)
- Chaoyue Zhang
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunke Shi
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Changzhi Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shivon Mirza Sudesh
- Faculty of Medicine, St. George University of London, London, UK
- University of Nicosia Medical School, University of Nicosia, Nicosia, Cyprus
| | - Zhao Hu
- Department of Geriatric Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Qi Liu
- Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Yiming Ma
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ao Shi
- Faculty of Medicine, St. George University of London, London, UK.
- University of Nicosia Medical School, University of Nicosia, Nicosia, Cyprus.
| | - Hongyan Cai
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Cai L, Tan Y, Watson S, Wintergerst K. Diabetic cardiomyopathy - Zinc preventive and therapeutic potentials by its anti-oxidative stress and sensitizing insulin signaling pathways. Toxicol Appl Pharmacol 2023; 477:116694. [PMID: 37739320 PMCID: PMC10616760 DOI: 10.1016/j.taap.2023.116694] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Oxidative stress and insulin resistance are two key mechanisms for the development of diabetic cardiomyopathy (DCM, cardiac remodeling and dysfunction). In this review, we discussed how zinc and metallothionein (MT) protect the heart from type 1 or type 2 diabetes (T1D or T2D) through its anti-oxidative function and insulin-mediated PI3K/Akt signaling activation. Both T1D and T2D-induced DCM, shown by cardiac structural remodeling and dysfunction, in wild-type mice, but not in cardiomyocyte-specific overexpressing MT mice. In contrast, mice with global MT gene deletion were more susceptible to the development of DCM. When we used zinc to treat mice with either T1D or T2D, cardiac remodeling and dysfunction were significantly prevented along with increased cardiac MT expression. To support the role of zinc homeostasis in insulin signaling pathways, treatment of diabetic mice with zinc showed the preservation of phosphorylation levels of insulin-mediated glucose metabolism-related Akt2 and GSK-3β and even rescued cardiac pathogenesis induced by global deletion of Akt2 gene in a MT-dependent manner. These results suggest the protection by zinc from DCM is through both the induction of MT and sensitization of insulin signaling. Combined our own and other works, this review comprehensively summarized the roles of zinc homeostasis in the development and progression of DCM and its therapeutic implications. At the end, we provided pre-clinical and clinical evidence for the preventive and therapeutic potential of zinc supplementation through its anti-oxidative stress and sensitizing insulin signaling actions. Understanding the intricate connections between zinc and DCM provides insights for the future interventional approaches.
Collapse
Affiliation(s)
- Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America; Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Sara Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| | - Kupper Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| |
Collapse
|
4
|
Zhang H, Shi C, Yang L, Zhang N, Li G, Zhou Z, Gao Y, Liu D, Xu L, Fan Z. Quantification of Early Diffuse Myocardial Fibrosis Through 7.0 T Cardiac Magnetic Resonance T1 Mapping in a Type 1 Diabetic Mellitus Mouse Model. J Magn Reson Imaging 2023; 57:167-177. [PMID: 35436040 DOI: 10.1002/jmri.28207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Diffuse myocardial interstitial fibrosis (DMIF) is a key factor for heart failure (HF) in diabetic cardiomyopathy. MRI T1-mapping technique can quantitatively evaluate DMIF. PURPOSE To evaluate of early DMIF in a type 1 diabetes mellitus (T1DM) mouse model through 7.0 T MRI T1 mapping. STUDY TYPE Prospective. ANIMAL MODEL A total of 50 8-week-old C57Bl/6J male mice were divided into control (n = 20) and T1DM (n = 30) groups. FIELD STRENGTH/SEQUENCE A 7.0 T small animal MRI; gradient echo Look-Locker inversion recovery T1-mapping sequence; cine MRI. Scans were acquired in control and T1DM mice every 4 weeks until 24 weeks. ASSESSMENT End-diastolic volume (EDV), end-systolic volume (ESV), ejection fraction (EF), left ventricle (LV) mass, fractional shortening (FS), and E/A ratio. They were evaluated through echocardiography and cine MRI. The extracellular volume fraction (ECV) was calculated. Sirius Red staining was performed and calculated collagen volume fraction (CVF). STATISTICAL TESTS Differences in ECV and CVF between two groups were analyzed using one-way analysis of variance. The correlation between ECV and CVF was assessed using Pearson's correlations. RESULTS Compared with the control group, a progressive decrease in FS, EF, and E/A ratio was observed in the T1DM group. Both ECV and CVF values gradually increased during diabetes progression. A significant increase in ECV and CVF values was observed at 12 weeks (ECV: 32.5% ± 1.6% vs. 28.1% ± 1.8%; CVF: 6.9% ± 1.8% vs. 3.3% ± 1.1%). ECV showed a strong correlation with CVF (r = 0.856). DATA CONCLUSION ECV is an accurate and feasible imaging marker that can be used to quantitatively assess DMIF changes over time in T1DM mice. ECV has potential to accurately detect DMIF in the early stage and may be a useful imaging tool to assess the need for early intervention in T1DM mice. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Hongkai Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Chunyan Shi
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Lin Yang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Nan Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Guoqi Li
- Beijing Institute of Heart, Lung & Vascular Diseases, The Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Zhen Zhou
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Yifeng Gao
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Dongting Liu
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Lei Xu
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| | - Zhanming Fan
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, China, 100029
| |
Collapse
|
5
|
Mladenov M, Bogdanov J, Bogdanov B, Hadzi-Petrushev N, Kamkin A, Stojchevski R, Avtanski D. Efficacy of the monocarbonyl curcumin analog C66 in the reduction of diabetes-associated cardiovascular and kidney complications. Mol Med 2022; 28:129. [PMID: 36316651 PMCID: PMC9620630 DOI: 10.1186/s10020-022-00559-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Curcumin is a polyphenolic compound derived from turmeric that has potential beneficial properties for cardiovascular and renal diseases and is relatively safe and inexpensive. However, the application of curcumin is rather problematic due to its chemical instability and low bioavailability. The experimental results showed improved chemical stability and potent pharmacokinetics of one of its analogs - (2E,6E)-2,6-bis[(2-trifluoromethyl)benzylidene]cyclohexanone (C66). There are several advantages of C66, like its synthetic accessibility, structural simplicity, improved chemical stability (in vitro and in vivo), presence of two reactive electrophilic centers, and good electron-accepting capacity. Considering these characteristics, we reviewed the literature on the application of C66 in resolving diabetes-associated cardiovascular and renal complications in animal models. We also summarized the mechanisms by which C66 is preventing the release of pro-oxidative and pro-inflammatory molecules in the priming and in activation stage of cardiomyopathy, renal fibrosis, and diabetic nephropathy. The cardiovascular protective effect of C66 against diabetes-induced oxidative damage is Nrf2 mediated but mainly dependent on JNK2. In general, C66 causes inhibition of JNK2, which reduces cardiac inflammation, fibrosis, oxidative stress, and apoptosis in the settings of diabetic cardiomyopathy. C66 exerts a powerful antifibrotic effect by reducing inflammation-related factors (MCP-1, NF-κB, TNF-α, IL-1β, COX-2, and CAV-1) and inducing the expression of anti-inflammatory factors (HO-1 and NEDD4), as well as targeting TGF-β/SMADs, MAPK/ERK, and PPAR-γ pathways in animal models of diabetic nephropathy. Based on the available evidence, C66 is becoming a promising drug candidate for improving cardiovascular and renal health.
Collapse
Affiliation(s)
- Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, Skopje, Macedonia
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street 1, Moscow, Russia
| | - Jane Bogdanov
- Faculty of Natural Sciences and Mathematics, Institute of Chemistry, Ss. Cyril and Methodius University in Skopje, Skopje, Macedonia
| | - Bogdan Bogdanov
- Faculty of Natural Sciences and Mathematics, Institute of Chemistry, Ss. Cyril and Methodius University in Skopje, Skopje, Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, Skopje, Macedonia
| | - Andre Kamkin
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street 1, Moscow, Russia
| | - Radoslav Stojchevski
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, Skopje, Macedonia
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, 110 E 59th Street, Suite 8B, Room 837, 10022, New York, NY, USA
| | - Dimiter Avtanski
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, 110 E 59th Street, Suite 8B, Room 837, 10022, New York, NY, USA.
- Feinstein Institutes for Medical Research, Manhasset, NY, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
6
|
Wang S, Tian C, Gao Z, Zhang B, Zhao L. Research status and trends of the diabetic cardiomyopathy in the past 10 years (2012–2021): A bibliometric analysis. Front Cardiovasc Med 2022; 9:1018841. [PMID: 36337893 PMCID: PMC9630656 DOI: 10.3389/fcvm.2022.1018841] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022] Open
Abstract
Background Diabetic cardiomyopathy is one of the most life-threatening diabetic complications. However, the previous studies only discuss a particular aspect or characteristic of DCM, the current state and trends were explored by limited research. We aimed to perform a systemically bibliometric study of DCM research progress status in the past decade, visualize the internal conceptual structure and potential associations, and further explore the prospective study trends. Methods Articles related to DCM published from January 2012 to December 2021 were collected in the Web of Science core collection (WoSCC) database on June 24, 2022. We exported all bibliographic records, including titles, abstracts, keywords, authorship, institutions, addresses, publishing sources, references, citation times, and year of publication. In addition, the journal Impact Factor and Hirsch index were obtained from the Journal Citation Report. We conducted the data screening, statistical analysis, and visualization via the Bibliometrix R package. VOS viewer software was employed to generate the collaboration network map among countries and institutions for better performance in visualization. Results In total, 1,887 original research articles from 2012 to 2021 were identified. The number of annual publications rapidly increased from 107 to 278, and a drastic increase in citation times was observed in 2017–2019. As for global contributions, the United States was the most influential country with the highest international collaboration, while China was the most productive country. Professor Cai Lu was the most prolific author. Shandong University published the most articles. Cardiovascular Diabetology journal released the most DCM-related articles. “Metabolic Stress-induced Activation of FoxO1 Triggers Diabetic Cardiomyopathy in Mice” Battiprolu PK et al., J Clin Invest, 2012. was the most top-cited article regarding local citations. The top three keywords in terms of frequency were apoptosis, oxidative stress, and fibrosis. The analysis of future topic trends indicated that “Forkhead box protein O1,” “Heart failure with preserved ejection fraction,” “Dapagliflozin,” “Thioredoxin,” “Mitochondria dysfunction,” “Glucose,” “Pyroptosis,” “Cardiac fibroblast” and “Long non-coding RNA” could be promising hotspots. Conclusion This study provides meaningful insights into DCM, which is expected to assist cardiologists and endocrinologists in exploring frontiers and future research directions in the domain through a refined and concise summary.
Collapse
Affiliation(s)
- Sicheng Wang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanxi Tian
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zezheng Gao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Boxun Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Boxun Zhang,
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Linhua Zhao,
| |
Collapse
|
7
|
Role of Oxidative Stress in Diabetic Cardiomyopathy. Antioxidants (Basel) 2022; 11:antiox11040784. [PMID: 35453469 PMCID: PMC9030255 DOI: 10.3390/antiox11040784] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes is a redox disease. Oxidative stress and chronic inflammation induce a switch of metabolic homeostatic set points, leading to glucose intolerance. Several diabetes-specific mechanisms contribute to prominent oxidative distress in the heart, resulting in the development of diabetic cardiomyopathy. Mitochondrial overproduction of reactive oxygen species in diabetic subjects is not only caused by intracellular hyperglycemia in the microvasculature but is also the result of increased fatty oxidation and lipotoxicity in cardiomyocytes. Mitochondrial overproduction of superoxide anion radicals induces, via inhibition of glyceraldehyde 3-phosphate dehydrogenase, an increased polyol pathway flux, increased formation of advanced glycation end-products (AGE) and activation of the receptor for AGE (RAGE), activation of protein kinase C isoforms, and an increased hexosamine pathway flux. These pathways not only directly contribute to diabetic cardiomyopathy but are themselves a source of additional reactive oxygen species. Reactive oxygen species and oxidative distress lead to cell dysfunction and cellular injury not only via protein oxidation, lipid peroxidation, DNA damage, and oxidative changes in microRNAs but also via activation of stress-sensitive pathways and redox regulation. Investigations in animal models of diabetic cardiomyopathy have consistently demonstrated that increased expression of the primary antioxidant enzymes attenuates myocardial pathology and improves cardiac function.
Collapse
|
8
|
Roberts AM, Moulana NZ, Jagadapillai R, Cai L, Gozal E. Intravital assessment of precapillary pulmonary arterioles of type 1 diabetic mice shows oxidative damage and increased tone in response to NOS inhibition. J Appl Physiol (1985) 2021; 131:1552-1564. [PMID: 34590907 PMCID: PMC11961051 DOI: 10.1152/japplphysiol.00395.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/07/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022] Open
Abstract
Microvascular dilation, important for peripheral tissue glucose distribution, also modulates alveolar perfusion and is inhibited by loss of bioavailable nitric oxide (NO) in diabetes mellitus (DM). We hypothesized that DM-induced oxidative stress decreases bioavailable NO and pulmonary precapillary arteriolar diameter, causing endothelial injury. We examined subpleural pulmonary arterioles after acute NO synthase (NOS) inhibition with NG-nitro-l-arginine methyl ester (l-NAME) in streptozotocin (STZ)- and saline (CTRL)-treated C57BL/6J mice. Microvascular changes were assessed by intravital microscopy in the right lung of anesthetized mice with open chest and ventilated lungs. Arteriolar tone in pulmonary arterioles (27.2-48.7 µm diameter) increased in CTRL mice (18.0 ± 11% constriction, P = 0.034, n = 5) but decreased in STZ mice (13.6 ± 7.5% dilation, P = 0.009, n = 5) after l-NAME. Lung tissue dihydroethidium (DHE) fluorescence (superoxide), inducible NOS expression, and protein nitrosylation (3-nitrotyrosine) increased in STZ mice and correlated with increased glucose levels (103.8 ± 8.8 mg/dL). Fluorescently labeled fibrinogen administration and fibrinogen immunostaining showed fibrinogen adhesion, indicating endothelial injury in STZ mice. In CTRL mice, vasoconstriction to l-NAME was likely due to the loss of bioavailable NO. Vasodilation in STZ mice may be due to decreased formation of a vasoconstrictor or emergence of a vasodilator. These findings provide novel evidence that DM targets the pulmonary microcirculation and that decreased NO bioavailability and increased precapillary arteriolar tone could potentially lead to ventilation-perfusion abnormalities, exacerbating systemic DM complications.NEW & NOTEWORTHY Diabetes pulmonary and microvascular consequences are well recognized but have not been characterized. We assessed lung microvascular changes in a live anesthetized mouse model of type 1 diabetes, using a novel intravital microscopy technique. Our results show new evidence that a diabetes-induced decrease in lung nitric oxide bioavailability underlies oxidative damage, enhanced platelet activation, and endothelial injury causing pulmonary microvascular dysfunction and altered vasoreactivity. These findings could provide novel strategies to prevent or reverse diabetes systemic consequences.
Collapse
Affiliation(s)
- Andrew M Roberts
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| | - Nayeem Z Moulana
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Rekha Jagadapillai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| | - Evelyne Gozal
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
9
|
Salvatore T, Pafundi PC, Galiero R, Albanese G, Di Martino A, Caturano A, Vetrano E, Rinaldi L, Sasso FC. The Diabetic Cardiomyopathy: The Contributing Pathophysiological Mechanisms. Front Med (Lausanne) 2021; 8:695792. [PMID: 34277669 PMCID: PMC8279779 DOI: 10.3389/fmed.2021.695792] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals with diabetes mellitus (DM) disclose a higher incidence and a poorer prognosis of heart failure (HF) than non-diabetic people, even in the absence of other HF risk factors. The adverse impact of diabetes on HF likely reflects an underlying “diabetic cardiomyopathy” (DM–CMP), which may by exacerbated by left ventricular hypertrophy and coronary artery disease (CAD). The pathogenesis of DM-CMP has been a hot topic of research since its first description and is still under active investigation, as a complex interplay among multiple mechanisms may play a role at systemic, myocardial, and cellular/molecular levels. Among these, metabolic abnormalities such as lipotoxicity and glucotoxicity, mitochondrial damage and dysfunction, oxidative stress, abnormal calcium signaling, inflammation, epigenetic factors, and others. These disturbances predispose the diabetic heart to extracellular remodeling and hypertrophy, thus leading to left ventricular diastolic and systolic dysfunction. This Review aims to outline the major pathophysiological changes and the underlying mechanisms leading to myocardial remodeling and cardiac functional derangement in DM-CMP.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
10
|
Byrne NJ, Rajasekaran NS, Abel ED, Bugger H. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radic Biol Med 2021; 169:317-342. [PMID: 33910093 PMCID: PMC8285002 DOI: 10.1016/j.freeradbiomed.2021.03.046] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Even in the absence of coronary artery disease and hypertension, diabetes mellitus (DM) may increase the risk for heart failure development. This risk evolves from functional and structural alterations induced by diabetes in the heart, a cardiac entity termed diabetic cardiomyopathy (DbCM). Oxidative stress, defined as the imbalance of reactive oxygen species (ROS) has been increasingly proposed to contribute to the development of DbCM. There are several sources of ROS production including the mitochondria, NAD(P)H oxidase, xanthine oxidase, and uncoupled nitric oxide synthase. Overproduction of ROS in DbCM is thought to be counterbalanced by elevated antioxidant defense enzymes such as catalase and superoxide dismutase. Excess ROS in the cardiomyocyte results in further ROS production, mitochondrial DNA damage, lipid peroxidation, post-translational modifications of proteins and ultimately cell death and cardiac dysfunction. Furthermore, ROS modulates transcription factors responsible for expression of antioxidant enzymes. Lastly, evidence exists that several pharmacological agents may convey cardiovascular benefit by antioxidant mechanisms. As such, increasing our understanding of the pathways that lead to increased ROS production and impaired antioxidant defense may enable the development of therapeutic strategies against the progression of DbCM. Herein, we review the current knowledge about causes and consequences of ROS in DbCM, as well as the therapeutic potential and strategies of targeting oxidative stress in the diabetic heart.
Collapse
Affiliation(s)
- Nikole J Byrne
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
11
|
Kumar V, Singh J, Bala K, Singh J. Association of Metallothionein 1A gene polymorphisms at rs11640851 and rs8052394 with risk of type 2 diabetes mellitus in Indian population. Meta Gene 2021. [DOI: 10.1016/j.mgene.2021.100862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
12
|
Xiao M, Tang Y, Wang S, Wang J, Wang J, Guo Y, Zhang J, Gu J. The Role of Fibroblast Growth Factor 21 in Diabetic Cardiovascular Complications and Related Epigenetic Mechanisms. Front Endocrinol (Lausanne) 2021; 12:598008. [PMID: 34349728 PMCID: PMC8326758 DOI: 10.3389/fendo.2021.598008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 06/17/2021] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21), is an emerging metabolic regulator mediates multiple beneficial effects in the treatment of metabolic disorders and related complications. Recent studies showed that FGF21 acts as an important inhibitor in the onset and progression of cardiovascular complications of diabetes mellitus (DM). Furthermore, evidences discussed so far demonstrate that epigenetic modifications exert a crucial role in the initiation and development of DM-related cardiovascular complications. Thus, epigenetic modifications may involve in the function of FGF21 on DM-induced cardiovascular complications. Therefore, this review mainly interprets and delineates the recent advances of role of FGF21 in DM cardiovascular complications. Then, the possible changes of epigenetics related to the role of FGF21 on DM-induced cardiovascular complications are discussed. Thus, this article not only implies deeper understanding of the pathological mechanism of DM-related cardiovascular complications, but also provides the possible novel therapeutic strategy for DM-induced cardiovascular complications by targeting FGF21 and related epigenetic mechanism.
Collapse
Affiliation(s)
- Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Shudong Wang
- Department of Cardiology at the First Hospital of Jilin University, Changchun, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People’s Hospital of Liaoning Province, Shenyang, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Junlian Gu,
| |
Collapse
|
13
|
Rajendran NK, Houreld NN, Abrahamse H. Photobiomodulation reduces oxidative stress in diabetic wounded fibroblast cells by inhibiting the FOXO1 signaling pathway. J Cell Commun Signal 2020; 15:195-206. [PMID: 33052534 DOI: 10.1007/s12079-020-00588-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
This study aimed to elucidate the underlying molecular mechanism of photobiomodulation (PBM) in attenuating oxidative stress in diabetic wounded fibroblast cells. Cell models were exposed to PBM at a wavelength of 660 nm (fluence of 5 J/cm2, and power density of 11.2 mW/cm2) or 830 nm (fluence of 5 J/cm2, and power density of 10.3 mW/cm2). Non-irradiated cell models were used as controls. Cellular migration was determined at regular time intervals (0, 12, 24 and 48 h) using inverted light microscopy. Cell viability was determined by the Trypan blue exclusion assay. The levels of enzymic antioxidants superoxide dismutase (SOD), catalase (CAT), and heme oxygenase (HMOX1) were determined by the enzyme linked immunosorbent assay (ELISA). The alteration in the levels of AKT and FOXO1 was determined by immunofluorescence and western blotting. Upon PBM treatment, elevated oxidative stress was reversed in diabetic and diabetic wounded fibroblast cells. The reduced oxidative stress was represented by decreased FOXO1 levels and increased levels of SOD, CAT and HMOX1. This might be due to the activation of the AKT signaling pathway. This study concluded that treatment with PBM progressed diabetic wound healing by attenuating oxidative stress through inhibition of the FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Naresh Kumar Rajendran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, 2028, South Africa.
| | - Nicolette Nadene Houreld
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, 2028, South Africa
| |
Collapse
|
14
|
Tan Y, Zhang Z, Zheng C, Wintergerst KA, Keller BB, Cai L. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol 2020; 17:585-607. [PMID: 32080423 PMCID: PMC7849055 DOI: 10.1038/s41569-020-0339-2] [Citation(s) in RCA: 446] [Impact Index Per Article: 89.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The pathogenesis and clinical features of diabetic cardiomyopathy have been well-studied in the past decade, but effective approaches to prevent and treat this disease are limited. Diabetic cardiomyopathy occurs as a result of the dysregulated glucose and lipid metabolism associated with diabetes mellitus, which leads to increased oxidative stress and the activation of multiple inflammatory pathways that mediate cellular and extracellular injury, pathological cardiac remodelling, and diastolic and systolic dysfunction. Preclinical studies in animal models of diabetes have identified multiple intracellular pathways involved in the pathogenesis of diabetic cardiomyopathy and potential cardioprotective strategies to prevent and treat the disease, including antifibrotic agents, anti-inflammatory agents and antioxidants. Some of these interventions have been tested in clinical trials and have shown favourable initial results. In this Review, we discuss the mechanisms underlying the development of diabetic cardiomyopathy and heart failure in type 1 and type 2 diabetes mellitus, and we summarize the evidence from preclinical and clinical studies that might provide guidance for the development of targeted strategies. We also highlight some of the novel pharmacological therapeutic strategies for the treatment and prevention of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- The Second Affiliated Hospital Center of Chinese-American Research Institute for Diabetic Complications, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA
- Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Bradley B Keller
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
15
|
Weng W, Ge T, Wang Y, He L, Liu T, Wang W, Zheng Z, Yu L, Zhang C, Lu X. Therapeutic Effects of Fibroblast Growth Factor-21 on Diabetic Nephropathy and the Possible Mechanism in Type 1 Diabetes Mellitus Mice. Diabetes Metab J 2020; 44:566-580. [PMID: 32431116 PMCID: PMC7453991 DOI: 10.4093/dmj.2019.0089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/28/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) has been only reported to prevent type 1 diabetic nephropathy (DN) in the streptozotocin-induced type 1 diabetes mellitus (T1DM) mouse model. However, the FVB (Cg)-Tg (Cryaa-Tag, Ins2-CALM1) 26OVE/PneJ (OVE26) transgenic mouse is a widely recommended mouse model to recapture the most important features of T1DM nephropathy that often occurs in diabetic patients. In addition, most previous studies focused on exploring the preventive effect of FGF21 on the development of DN. However, in clinic, development of therapeutic strategy has much more realistic value compared with preventive strategy since the onset time of DN is difficult to be accurately predicted. Therefore, in the present study OVE26 mice were used to investigate the potential therapeutic effects of FGF21 on DN. METHODS Four-month-old female OVE26 mice were intraperitoneally treated with recombinant FGF21 at a dose of 100 μg/kg/day for 3 months. The diabetic and non-diabetic control mice were treated with phosphate-buffered saline at the same volume. Renal functions, pathological changes, inflammation, apoptosis, oxidative stress and fibrosis were examined in mice of all groups. RESULTS The results showed that severe renal dysfunction, morphological changes, inflammation, apoptosis, and fibrosis were observed in OVE26 mice. However, all the renal abnormalities above in OVE26 mice were significantly attenuated by 3-month FGF21 treatment associated with improvement of renal adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) activity and sirtuin 1 (SIRT1) expression. CONCLUSION Therefore, this study demonstrated that FGF21 might exert therapeutic effects on DN through AMPK-SIRT1 pathway.
Collapse
Affiliation(s)
- Wenya Weng
- Ruian Center of the Chinese-American Institute for Diabetic Complications, the Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tingwen Ge
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| | - Yi Wang
- Biological Engineering Department, School of Life Science, Anhui Medical University, Hefei, China
| | - Lulu He
- The Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Tinghao Liu
- The Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Wanning Wang
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| | - Zongyu Zheng
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| | - Lechu Yu
- Ruian Center of the Chinese-American Institute for Diabetic Complications, the Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chi Zhang
- Ruian Center of the Chinese-American Institute for Diabetic Complications, the Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xuemian Lu
- Ruian Center of the Chinese-American Institute for Diabetic Complications, the Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
16
|
Wang K, Dai X, He J, Yan X, Yang C, Fan X, Sun S, Chen J, Xu J, Deng Z, Fan J, Yuan X, Liu H, Carlson EC, Shen F, Wintergerst KA, Conklin DJ, Epstein PN, Lu C, Tan Y. Endothelial Overexpression of Metallothionein Prevents Diabetes-Induced Impairment in Ischemia Angiogenesis Through Preservation of HIF-1α/SDF-1/VEGF Signaling in Endothelial Progenitor Cells. Diabetes 2020; 69:1779-1792. [PMID: 32404351 PMCID: PMC7519474 DOI: 10.2337/db19-0829] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 05/09/2020] [Indexed: 12/13/2022]
Abstract
Diabetes-induced oxidative stress is one of the major contributors to dysfunction of endothelial progenitor cells (EPCs) and impaired endothelial regeneration. Thus, we tested whether increasing antioxidant protein metallothionein (MT) in EPCs promotes angiogenesis in a hind limb ischemia (HLI) model in endothelial MT transgenic (JTMT) mice with high-fat diet- and streptozocin-induced diabetes. Compared with littermate wild-type (WT) diabetic mice, JTMT diabetic mice had improved blood flow recovery and angiogenesis after HLI. Similarly, transplantation of JTMT bone marrow-derived mononuclear cells (BM-MNCs) stimulated greater blood flow recovery in db/db mice with HLI than did WT BM-MNCs. The improved recovery was associated with augmented EPC mobilization and angiogenic function. Further, cultured EPCs from patients with diabetes exhibited decreased MT expression, increased cell apoptosis, and impaired tube formation, while cultured JTMT EPCs had enhanced cell survival, migration, and tube formation in hypoxic/hyperglycemic conditions compared with WT EPCs. Mechanistically, MT overexpression enhanced hypoxia-inducible factor 1α (HIF-1α), stromal cell-derived factor (SDF-1), and vascular endothelial growth factor (VEGF) expression and reduced oxidative stress in ischemic tissues. MT's pro-EPC effects were abrogated by siRNA knockdown of HIF-1α without affecting its antioxidant action. These results indicate that endothelial MT overexpression is sufficient to protect against diabetes-induced impairment of angiogenesis by promoting EPC function, most likely through upregulation of HIF-1α/SDF-1/VEGF signaling and reducing oxidative stress.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Survival/genetics
- Cell Survival/physiology
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/prevention & control
- Endothelial Progenitor Cells/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Hindlimb/pathology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Ischemia/genetics
- Ischemia/metabolism
- Leukocytes, Mononuclear/metabolism
- Male
- Metallothionein/genetics
- Metallothionein/metabolism
- Mice
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- Oxidative Stress/genetics
- Oxidative Stress/physiology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Kai Wang
- Department of Pediatrics, Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Xiaozhen Dai
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Junhong He
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaoqing Yan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chengkui Yang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Xia Fan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shiyue Sun
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Chen
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Jianxiang Xu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Zhongbin Deng
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY
| | - Jiawei Fan
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Xiaohuan Yuan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China
| | - Hairong Liu
- Experimental Research Center, the First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, China
| | - Edward C Carlson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND
| | - Feixia Shen
- Department of Pediatrics, Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children's Medical Group, Louisville, KY
- Wendy Novak Diabetes Center, Louisville, KY
| | - Daniel J Conklin
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY
- Diabetes and Obesity Center, University of Louisville, Louisville, KY
| | - Paul N Epstein
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Wendy Novak Diabetes Center, Louisville, KY
| | - Chaosheng Lu
- Department of Pediatrics, Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Wendy Novak Diabetes Center, Louisville, KY
| |
Collapse
|
17
|
Okesola MA, Ojo OA, Onikanni SA, Ajiboye BO, Oyinloye BE, Agboinghale PE, Kappo AP. Ameliorative effect of Gongronema latifolium leaf extract on alloxan-induced diabetic cardiomyopathy in Wistar rats model. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s00580-020-03134-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Lin X, Jagadapillai R, Cai J, Cai L, Shao G, Gozal E. Metallothionein induction attenuates the progression of lung injury in mice exposed to long-term intermittent hypoxia. Inflamm Res 2020; 69:15-26. [PMID: 31707449 DOI: 10.1007/s00011-019-01287-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 09/19/2019] [Accepted: 09/24/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Intermittent hypoxia (IH), a hallmark of obstructive sleep apnea (OSA), is prevalent in older adults and associated with inflammation. We previously showed that IH induces renal fibrosis and cardiomyopathy and hypothesized that lung inflammatory changes may underlie deficits in pulmonary function in OSA. METHODS Pulmonary inflammatory and oxidative markers were assessed in metallothionein KO (MT-KO) mice and WT 129S1 controls exposed to IH or to normoxia for 8 weeks. RESULTS MT expression increased at 3 days in WT, falling back at 1 week. Pro-fibrotic markers CTGF and PAI-1 were unchanged in WT, but increased at 3 or 8 weeks, with enhanced Sirius Red staining at 8 weeks, in IH-exposed MT-KO. Cellular infiltration, TNF-α and IL-6 increased earlier in IH-exposed MT-KO than in WT. Oxidative markers, 3-nitrotyrosine and 4-hydroxynonenal increased in both but persisted in MT-KO. Antioxidant Nrf2, HO-1 and NQO1, increased at 3 days in WT mice and at 8 weeks IH in MT-KO. While early Nrf2 induction required MT, its later increase at 8 weeks in MT-KO was independent from MT. CONCLUSIONS We conclude that early MT and antioxidant gene response protects from fibrotic changes in long-term IH-exposed mouse lung. Without this response, pulmonary fibrosis may develop with longer IH exposure.
Collapse
Affiliation(s)
- Xingyu Lin
- Department of Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
- PRI, Departments of Pediatrics, University of Louisville, 570, S. Preston St., Ste 321, Louisville, 40202, USA
| | - Rekha Jagadapillai
- PRI, Departments of Pediatrics, University of Louisville, 570, S. Preston St., Ste 321, Louisville, 40202, USA
| | - Jun Cai
- PRI, Departments of Pediatrics, University of Louisville, 570, S. Preston St., Ste 321, Louisville, 40202, USA
| | - Lu Cai
- PRI, Departments of Pediatrics, University of Louisville, 570, S. Preston St., Ste 321, Louisville, 40202, USA
- Pharmacology and Toxicology, University of Louisville, Louisville, 40202, USA
| | - Guoguang Shao
- Department of Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.
| | - Evelyne Gozal
- PRI, Departments of Pediatrics, University of Louisville, 570, S. Preston St., Ste 321, Louisville, 40202, USA.
- Pharmacology and Toxicology, University of Louisville, Louisville, 40202, USA.
- Physiology School of Medicine, University of Louisville, Louisville, 40202, USA.
| |
Collapse
|
19
|
Natarajan J, Sanapalli BKR, Bano M, Singh SK, Gulati M, Karri VVSR. Nanostructured Lipid Carriers of Pioglitazone Loaded Collagen/Chitosan Composite Scaffold for Diabetic Wound Healing. Adv Wound Care (New Rochelle) 2019; 8:499-513. [PMID: 31737408 PMCID: PMC6855294 DOI: 10.1089/wound.2018.0831] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/20/2019] [Indexed: 12/26/2022] Open
Abstract
Diabetic wound is a major problem that often needs amputation of the concerned organ in patients suffering from diabetes. In diabetes, the prolonged phase of inflammation obstructs the further phases of healing which, in turn, lead to improper healing of the wounds in diabetes. Pioglitazone (Pio) hydrochloride is an antidiabetic drug with reported anti-inflammatory properties. The aim of this study was to develop a Pio-nanostructured lipid carrier (Pio-NLC)-loaded collagen/chitosan (COL-CS) scaffold and evaluate its healing ability in diabetic wounds. The results of characterization of composite scaffolds reveal that cross-linked scaffolds possess optimum porosity, low matrix degradation, and sustained drug release compared with noncross-linked scaffolds. The in vitro studies reveal that the Pio-NLC-COL-CS scaffold was biocompatible and enhanced cell growth compared with control and NLC-COL-CS. Using the streptozotocin-induced diabetic wound model, significantly (p < 0.001) higher rates of wound contraction in Pio-NLC-COL-CS scaffold-treated group were observed in comparison with that in control and NLC-COL-CS-treated group. The enzyme-linked immunosorbent assay results indicate a significant (p < 0.001) decrease of matrix metalloproteinases-9 levels in the Pio-NLC-COL-CS-treated group compared with those in control group. Use of nanostructured lipid carrier (Pio-NLC-COL-CS) scaffold can prove to be a promising strategy for local treatment for diabetic wounds.
Collapse
Affiliation(s)
- Jawahar Natarajan
- Department of Pharmaceutics, JSS College of Pharmacy, Ooty, JSS Academy of Higher Education and Research, India
| | | | - Mehjabeen Bano
- Department of Pharmaceutics, JSS College of Pharmacy, Ooty, JSS Academy of Higher Education and Research, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Monica Gulati
- Faculty of Applied Medical Sciences, Lovely Professional University, Phagwara, India
| | | |
Collapse
|
20
|
Wang J, Wang S, Wang W, Chen J, Zhang Z, Zheng Q, Liu Q, Cai L. Protection against diabetic cardiomyopathy is achieved using a combination of sulforaphane and zinc in type 1 diabetic OVE26 mice. J Cell Mol Med 2019; 23:6319-6330. [PMID: 31270951 PMCID: PMC6714218 DOI: 10.1111/jcmm.14520] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/20/2019] [Accepted: 06/15/2019] [Indexed: 12/17/2022] Open
Abstract
Sulforaphane (SFN) can effectively induce nuclear factor E2-related factor 2 (Nrf2), and zinc (Zn) can effectively induce metallothionein (MT), both of which have been shown to protect against diabetic cardiomyopathy (DCM). However, it is unclear whether combined treatment with SFN and Zn offers better cardiac protection than either one alone. Here, we treated 5-week-old OVE mice that spontaneously develop type 1 diabetes with SFN and/or Zn for 18 weeks. Cardiac dysfunction, by echocardiography, and pathological alterations and remodelling, shown by cardiac hypertrophy, fibrosis, inflammation and oxidative damage, examined by histopathology, Western blotting and real-time PCR, were observed in OVE mice. All these dysfunction and pathological abnormalities seen in OVE mice were attenuated in OVE mice with treatment of either SFN, Zn or SFN/Zn, and the combined treatment with SFN/Zn was better than single treatments at ameliorating DCM. In addition, combined SFN and Zn treatment increased Nrf2 function and MT expression in the heart of OVE mice to a greater extent than SFN or Zn alone. This indicates that the dual activation of Nrf2 and MT by combined treatment with SFN and Zn may be more effective than monotherapy at preventing the development of DCM via complementary, additive mechanisms.
Collapse
Affiliation(s)
- Jiqun Wang
- The Center of Cardiovascular DiseasesThe First Hospital of Jilin UniversityChangchunChina
- Pediatric Research Institute, Department of PediatricsUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Shudong Wang
- The Center of Cardiovascular DiseasesThe First Hospital of Jilin UniversityChangchunChina
| | - Wanning Wang
- Pediatric Research Institute, Department of PediatricsUniversity of LouisvilleLouisvilleKentuckyUSA
- Department of NephrologyThe First Hospital of Jilin UniversityChangchunChina
| | - Jing Chen
- Department of OtolaryngologyStanford UniversityPalo AltoCaliforniaUSA
| | - Zhiguo Zhang
- The Center of Cardiovascular DiseasesThe First Hospital of Jilin UniversityChangchunChina
| | - Qi Zheng
- Department of Bioinformatics and BiostatisticsUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Quan Liu
- The Center of Cardiovascular DiseasesThe First Hospital of Jilin UniversityChangchunChina
| | - Lu Cai
- Pediatric Research Institute, Department of PediatricsUniversity of LouisvilleLouisvilleKentuckyUSA
- Departments of Radiation Oncology, Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKentuckyUSA
| |
Collapse
|
21
|
Ge T, Yu Y, Cui J, Cai L. The adaptive immune role of metallothioneins in the pathogenesis of diabetic cardiomyopathy: good or bad. Am J Physiol Heart Circ Physiol 2019; 317:H264-H275. [PMID: 31100011 DOI: 10.1152/ajpheart.00123.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetes is a metabolic disorder characterized by hyperglycemia, resulting in low-grade systemic inflammation. Diabetic cardiomyopathy (DCM) is a common complication among diabetic patients, and the mechanism underlying its induction of cardiac remodeling and dysfunction remains unclear. Numerous experimental and clinical studies have suggested that adaptive immunity, especially T lymphocyte-mediated immunity, plays a potentially important role in the pathogenesis of diabetes and DCM. Metallothioneins (MTs), cysteine-rich, metal-binding proteins, have antioxidant properties. Some potential mechanisms underlying the cardioprotective effects of MTs include the role of MTs in calcium regulation, zinc homeostasis, insulin sensitization, and antioxidant activity. Moreover, metal homeostasis, especially MT-regulated zinc homeostasis, is essential for immune function. This review discusses aberrant immune regulation in diabetic heart disease with respect to endothelial insulin resistance and the effects of hyperglycemia and hyperlipidemia on tissues and the different effects of intracellular and extracellular MTs on adaptive immunity. This review shows that intracellular MTs are involved in naïve T-cell activation and reduce regulatory T-cell (Treg) polarization, whereas extracellular MTs promote proliferation and survival in naïve T cells and Treg polarization but inhibit their activation, thus revealing potential therapeutic strategies targeting the regulation of immune cell function by MTs.
Collapse
Affiliation(s)
- Tingwen Ge
- Cancer Center, First Hospital of Jilin University , Changchun, Jilin , China
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Norton Health Care, Louisville, Kentucky
| | - Youxi Yu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Norton Health Care, Louisville, Kentucky
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jilin University , Changchun, Jilin , China
| | - Jiuwei Cui
- Cancer Center, First Hospital of Jilin University , Changchun, Jilin , China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Norton Health Care, Louisville, Kentucky
- Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville , Louisville, Kentucky
| |
Collapse
|
22
|
Ji K, Wang Y, Du L, Xu C, Liu Y, He N, Wang J, Liu Q. Research Progress on the Biological Effects of Low-Dose Radiation in China. Dose Response 2019; 17:1559325819833488. [PMID: 30833876 PMCID: PMC6393828 DOI: 10.1177/1559325819833488] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/19/2018] [Accepted: 12/29/2018] [Indexed: 01/07/2023] Open
Abstract
Human are exposed to ionizing radiation from natural and artificial sources, which consequently poses a possible risk to human health. However, accumulating evidence indicates that the biological effects of low-dose radiation (LDR) are different from those of high-dose radiation (HDR). Low-dose radiation–induced hormesis has been extensively observed in different biological systems, including immunological and hematopoietic systems. Adaptive responses in response to LDR that can induce cellular resistance to genotoxic effects from subsequent exposure to HDR have also been described and researched. Bystander effects, another type of biological effect induced by LDR, have been shown to widely occur in many cell types. Furthermore, the influence of LDR-induced biological effects on certain diseases, such as cancer and diabetes, has also attracted the interest of researchers. Many studies have suggested that LDR has the potential antitumor and antidiabetic complications effects. In addition, the researches on whether LDR could induce stochastic effects were also debated. Studies on the biological effects of LDR in China started in 1970s and considerable progress has been made since. In the present article, we provide an overview of the research progress on the biological effects of LDR in China.
Collapse
Affiliation(s)
- Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Yan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Chang Xu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Jinhan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| |
Collapse
|
23
|
Lighthouse JK, Burke RM, Velasquez LS, Dirkx RA, Aiezza A, Moravec CS, Alexis JD, Rosenberg A, Small EM. Exercise promotes a cardioprotective gene program in resident cardiac fibroblasts. JCI Insight 2019; 4:92098. [PMID: 30626739 DOI: 10.1172/jci.insight.92098] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
Exercise and heart disease both induce cardiac remodeling, but only disease causes fibrosis and compromises heart function. The cardioprotective benefits of exercise have been attributed to changes in cardiomyocyte physiology, but the impact of exercise on cardiac fibroblasts (CFs) is unknown. Here, RNA-sequencing reveals rapid divergence of CF transcriptional programs during exercise and disease. Among the differentially expressed programs, NRF2-dependent antioxidant genes - including metallothioneins (Mt1 and Mt2) - are induced in CFs during exercise and suppressed by TGF-β/p38 signaling in disease. In vivo, mice lacking Mt1/2 exhibit signs of cardiac dysfunction in exercise, including cardiac fibrosis, vascular rarefaction, and functional decline. Mechanistically, exogenous MTs derived from fibroblasts are taken up by cultured cardiomyocytes, reducing oxidative damage-dependent cell death. Importantly, suppression of MT expression is conserved in human heart failure. Taken together, this study defines the acute transcriptional response of CFs to exercise and disease and reveals a cardioprotective mechanism that is lost in disease.
Collapse
Affiliation(s)
- Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Ryan M Burke
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Lissette S Velasquez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Ronald A Dirkx
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Alessandro Aiezza
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | - Alex Rosenberg
- Department of Allergy, Immunology, and Rheumatology Research, and
| | - Eric M Small
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.,Department of Medicine.,Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
24
|
Sarkar A, Shukla SK, Alqatawni A, Kumar A, Addya S, Tsygankov AY, Rafiq K. The Role of Allograft Inflammatory Factor-1 in the Effects of Experimental Diabetes on B Cell Functions in the Heart. Front Cardiovasc Med 2018; 5:126. [PMID: 30258845 PMCID: PMC6145033 DOI: 10.3389/fcvm.2018.00126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/21/2018] [Indexed: 01/18/2023] Open
Abstract
Diabetes mellitus (DM) often causes chronic inflammation, hypertrophy, apoptosis and fibrosis in the heart and subsequently leads to myocardial remodeling, deteriorated cardiac function and heart failure. However, the etiology of the cardiac disease is unknown. Therefore, we assessed the gene expression in the left ventricle of diabetic and non-diabetic mice using Affymetrix microarray analysis. Allograft inflammatory factor-1 (AIF-1), one of the top downregulated B cell inflammatory genes, is associated with B cell functions in inflammatory responses. Real-time reverse transcriptase-polymerase chain reaction confirmed the Affymetrix data. The expression of CD19 and AIF-1 were downregulated in diabetic hearts as compared to control hearts. Using in vitro migration assay, we showed for the first time that AIF-1 is responsible for B cell migration as B cells migrated to GFP-AIF-1-transfected H9C2 cells compared to empty vector-transfected cells. Interestingly, overexpression of AIF-1 in diabetic mice prevented streptozotocin-induced cardiac dysfunction, inflammation and promoted B cell homing into the heart. Our results suggest that AIF-1 downregulation inhibited B cell homing into diabetic hearts, thus promoting inflammation that leads to the development of diabetic cardiomyopathy, and that overexpression of AIF-1 could be a novel treatment for this condition.
Collapse
Affiliation(s)
- Amrita Sarkar
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sanket K Shukla
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Aseel Alqatawni
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Anil Kumar
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sankar Addya
- Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexander Y Tsygankov
- Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Khadija Rafiq
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
25
|
Dharmarwardana M, Martins AF, Chen Z, Palacios PM, Nowak CM, Welch RP, Li S, Luzuriaga MA, Bleris L, Pierce BS, Sherry AD, Gassensmith JJ. Nitroxyl Modified Tobacco Mosaic Virus as a Metal-Free High-Relaxivity MRI and EPR Active Superoxide Sensor. Mol Pharm 2018; 15:2973-2983. [PMID: 29771534 PMCID: PMC6078806 DOI: 10.1021/acs.molpharmaceut.8b00262] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Superoxide overproduction is known to occur in multiple disease states requiring critical care; yet, noninvasive detection of superoxide in deep tissue remains a challenge. Herein, we report a metal-free magnetic resonance imaging (MRI) and electron paramagnetic resonance (EPR) active contrast agent prepared by "click conjugating" paramagnetic organic radical contrast agents (ORCAs) to the surface of tobacco mosaic virus (TMV). While ORCAs are known to be reduced in vivo to an MRI/EPR silent state, their oxidation is facilitated specifically by reactive oxygen species-in particular, superoxide-and are largely unaffected by peroxides and molecular oxygen. Unfortunately, single molecule ORCAs typically offer weak MRI contrast. In contrast, our data confirm that the macromolecular ORCA-TMV conjugates show marked enhancement for T1 contrast at low field (<3.0 T) and T2 contrast at high field (9.4 T). Additionally, we demonstrated that the unique topology of TMV allows for a "quenchless fluorescent" bimodal probe for concurrent fluorescence and MRI/EPR imaging, which was made possible by exploiting the unique inner and outer surface of the TMV nanoparticle. Finally, we show TMV-ORCAs do not respond to normal cellular respiration, minimizing the likelihood for background, yet still respond to enzymatically produced superoxide in complicated biological fluids like serum.
Collapse
Affiliation(s)
- Madushani Dharmarwardana
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - André F. Martins
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, USA
| | - Zhuo Chen
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Philip M. Palacios
- Department of Chemistry and Biochemistry, College of Sciences, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - Chance M. Nowak
- Department of Biological Sciences, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Raymond P. Welch
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Shaobo Li
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Michael A. Luzuriaga
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Leonidas Bleris
- Department of Biological Sciences, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| | - Brad S. Pierce
- Department of Chemistry and Biochemistry, College of Sciences, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - A. Dean Sherry
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, USA
| | - Jeremiah J. Gassensmith
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080-3021, USA
| |
Collapse
|
26
|
Xin Y, Bai Y, Jiang X, Zhou S, Wang Y, Wintergerst KA, Cui T, Ji H, Tan Y, Cai L. Sulforaphane prevents angiotensin II-induced cardiomyopathy by activation of Nrf2 via stimulating the Akt/GSK-3ß/Fyn pathway. Redox Biol 2018; 15:405-417. [PMID: 29353218 PMCID: PMC5975128 DOI: 10.1016/j.redox.2017.12.016] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/17/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023] Open
Abstract
AIMS Activation of nuclear factor erythroid 2-related factor 2 (Nrf2) by sulforaphane (SFN) protects from, and deletion of the Nrf2 gene exaggerates, diabetic cardiomyopathy. Angiotensin II (Ang II) plays a critical role in the development of diabetic cardiomyopathy. Therefore, whether SFN prevents Ang II-induced cardiomyopathy through activation of Nrf2 was examined using wild-type, global deletion of Nrf2 gene (Nrf2-KO) and cardiomyocyte-specific overexpression of Nrf2 gene (Nrf2-TG) mice. METHODS AND RESULTS Administration of a subpressor dose of Ang II to wild-type mice induced cardiac oxidative stress, inflammation, remodeling and dysfunction, all of which could be prevented by SFN treatment with Nrf2 up-regulation and activation. Nrf2-KO mice are susceptible, and Nrf2-TG mice are resistant, respectively, to Ang II-induced cardiomyopathy. Meanwhile, the ability of SFN to protect against Ang II-induced cardiac damage was lost in Nrf2-KO mice. Up-regulation and activation of Nrf2 by SFN is accompanied by activation of Akt, inhibition of glycogen synthase kinase (GSK)-3β, and accumulation of Fyn in nuclei. In vitro up-regulation of Nrf2 by SFN was abolished and nuclear Fyn accumulation was increased when cardiac cells were exposed to a PI3K inhibitor or GSK-3β-specific activator. CONCLUSION These results suggest that Nrf2 plays a central role in the prevention of Ang II-induced cardiomyopathy, and SFN prevents Ang II-induced cardiomyopathy partially via the Akt/GSK-3β/Fyn-mediated Nrf2 activation.
Collapse
Affiliation(s)
- Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences at the Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Research Institute, the Department of Pediatrics of the University of Louisville, Louisville 40202, USA
| | - Yang Bai
- Pediatrics Research Institute, the Department of Pediatrics of the University of Louisville, Louisville 40202, USA; First Hospital of Jilin University, Changchun 130021, China
| | - Xin Jiang
- Pediatrics Research Institute, the Department of Pediatrics of the University of Louisville, Louisville 40202, USA; First Hospital of Jilin University, Changchun 130021, China
| | - Shanshan Zhou
- Pediatrics Research Institute, the Department of Pediatrics of the University of Louisville, Louisville 40202, USA; First Hospital of Jilin University, Changchun 130021, China
| | - Yuehui Wang
- First Hospital of Jilin University, Changchun 130021, China
| | - Kupper A Wintergerst
- Division of Endocrinology of Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville 40202, USA; Wendy L. Novak Diabetes Care Center at the University of Louisville, Louisville 40202, USA
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia 29208, USA
| | - Honglei Ji
- First Hospital of Jilin University, Changchun 130021, China.
| | - Yi Tan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences at the Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Research Institute, the Department of Pediatrics of the University of Louisville, Louisville 40202, USA; Wendy L. Novak Diabetes Care Center at the University of Louisville, Louisville 40202, USA.
| | - Lu Cai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences at the Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Research Institute, the Department of Pediatrics of the University of Louisville, Louisville 40202, USA; Wendy L. Novak Diabetes Care Center at the University of Louisville, Louisville 40202, USA; Department of Pharmacology and Toxicology, the University of Louisville, Louisville 40202, USA
| |
Collapse
|
27
|
Xiao H, Wu C, Li P, Tang B. Simultaneous Fluorescence Visualization of Endoplasmic Reticulum Superoxide Anion and Polarity in Myocardial Cells and Tissue. Anal Chem 2018; 90:6081-6088. [DOI: 10.1021/acs.analchem.7b05440] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Haibin Xiao
- Collaborative Innovation Center of Functionalized
Probes for Chemical Imaging in Universities of Shandong, Key Laboratory
of Molecular and Nano Probes, Ministry of Education, College of Chemistry, Chemical Engineering and Materials Science, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Chuanchen Wu
- Collaborative Innovation Center of Functionalized
Probes for Chemical Imaging in Universities of Shandong, Key Laboratory
of Molecular and Nano Probes, Ministry of Education, College of Chemistry, Chemical Engineering and Materials Science, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Ping Li
- Collaborative Innovation Center of Functionalized
Probes for Chemical Imaging in Universities of Shandong, Key Laboratory
of Molecular and Nano Probes, Ministry of Education, College of Chemistry, Chemical Engineering and Materials Science, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Bo Tang
- Collaborative Innovation Center of Functionalized
Probes for Chemical Imaging in Universities of Shandong, Key Laboratory
of Molecular and Nano Probes, Ministry of Education, College of Chemistry, Chemical Engineering and Materials Science, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, People’s Republic of China
| |
Collapse
|
28
|
Wang G, Song X, Zhao L, Li Z, Liu B. Resveratrol Prevents Diabetic Cardiomyopathy by Increasing Nrf2 Expression and Transcriptional Activity. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2150218. [PMID: 29721501 PMCID: PMC5867593 DOI: 10.1155/2018/2150218] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This study investigated if resveratrol ameliorates diabetic cardiomyopathy by targeting associated oxidative stress mechanisms. METHOD Type 1 diabetes mellitus (DM) in FVB mice was induced by several intraperitoneal injections of a low dose of streptozotocin. Hyperglycemic and age-matched control mice were given resveratrol (10 mg/kg per day) for 1 month and subsequently monitored for an additional 6 months. Mice were assigned to four groups: control, resveratrol, DM, and DM/resveratrol. Cardiac function and blood pressure were assessed at 1, 3, and 6 months after DM induction. Oxidative damage and cardiac fibrosis were analyzed by histopathology, real-time PCR, and Western blot. RESULT Mice in the DM group exhibited increased blood glucose levels, cardiac dysfunction, and high blood pressure at 1, 3, and 6 months after DM induction. Resveratrol did not significantly affect blood glucose levels and blood pressure; however, resveratrol attenuated cardiac dysfunction and hypertrophy in DM mice. Resveratrol also reduced DM-induced fibrosis. In addition, DM mice hearts exhibited increased oxidative damage, as evidenced by elevated accumulation of 3-nitrotyrosine and 4-hydroxynonenal, which were both attenuated by resveratrol. Mechanistically, resveratrol increased NFE2-related factor 2 (Nrf2) expression and transcriptional activity, as well as Nrf2's downstream antioxidative targets. CONCLUSION We demonstrated that resveratrol prevents DM-induced cardiomyopathy, in part, by increasing Nrf2 expression and transcriptional activity.
Collapse
Affiliation(s)
- Guan Wang
- Department of Cardiology, The Second Hospital of Jilin University, Jilin University, Changchun 100032, China
| | - Xianjin Song
- Department of Cardiology, The Second Hospital of Jilin University, Jilin University, Changchun 100032, China
| | - Lei Zhao
- Department of Cardiology, The Second Hospital of Jilin University, Jilin University, Changchun 100032, China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Jilin University, Changchun 100032, China
| | - Bing Liu
- Department of Cardiology, The Second Hospital of Jilin University, Jilin University, Changchun 100032, China
| |
Collapse
|
29
|
Park Y, Zhang J, Cai L. Reappraisal of metallothionein: Clinical implications for patients with diabetes mellitus. J Diabetes 2018; 10:213-231. [PMID: 29072367 DOI: 10.1111/1753-0407.12620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 08/29/2017] [Accepted: 10/20/2017] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen and nitrogen species (ROS and RNS, respectively) are byproducts of cellular physiological processes of the metabolism of intermediary nutrients. Although physiological defense mechanisms readily convert these species into water or urea, an improper balance between their production and removal leads to oxidative stress (OS), which is harmful to cellular components. This OS may result in uncontrolled growth or, ultimately, cell death. In addition, ROS and RNS are closely related to the development of diabetes and its complications. Therefore, numerous researchers have proposed the development of strategies for the removal of ROS/RNS to prevent or treat diabetes and its complications. Some molecules that are synthesized in the body or obtained from food participate in the removal and neutralization of ROS and RNS. Metallothionein, a cysteine-rich protein, is a metal-binding protein that has a wide range of functions in cellular homeostasis and immunity. Metallothionein can be induced by a variety of conditions, including zinc supplementation, and plays a crucial role in mediating anti-OS, anti-apoptotic, detoxification, and anti-inflammatory effects. Metallothionein can modulate various stress-induced signaling pathways (mitogen-activated protein kinase, Wnt, nuclear factor-κB, phosphatidylinositol 3-kinase, sirtuin 1/AMP-activated protein kinase and fibroblast growth factor 21) to alleviate diabetes and diabetic complications. However, a deeper understanding of the functional, biochemical, and molecular characteristics of metallothionein is needed to bring about new opportunities for OS therapy. This review focuses on newly proposed functions of a metallothionein and their implications relevant to diabetes and its complications.
Collapse
Affiliation(s)
- Yongsoo Park
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- Hanyang University, College of Medicine and Engineering, Seoul, South Korea
| | - Jian Zhang
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- The Center of Cardiovascular Disorders, The First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Radiation Oncology, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
30
|
Gu J, Yan X, Dai X, Wang Y, Lin Q, Xiao J, Zhou S, Zhang J, Wang K, Zeng J, Xin Y, Barati MT, Zhang C, Bai Y, Li Y, Epstein PN, Wintergerst KA, Li X, Tan Y, Cai L. Metallothionein Preserves Akt2 Activity and Cardiac Function via Inhibiting TRB3 in Diabetic Hearts. Diabetes 2018; 67:507-517. [PMID: 29079702 PMCID: PMC5828458 DOI: 10.2337/db17-0219] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/20/2017] [Indexed: 12/17/2022]
Abstract
Cardiac insulin resistance is a key pathogenic factor for diabetic cardiomyopathy (DCM), but the mechanism remains largely unclear. We found that diabetic hearts exhibited decreased phosphorylation of total Akt and isoform Akt2 but not Akt1 in wild-type (WT) male FVB mice, which was accompanied by attenuation of Akt downstream glucose metabolic signal. All of these signal changes were not observed in metallothionein cardiac-specific transgenic (MT-TG) hearts. Furthermore, insulin-induced glucose metabolic signals were attenuated only in WT diabetic hearts. In addition, diabetic hearts exhibited increased Akt-negative regulator tribbles pseudokinase 3 (TRB3) expression only in WT mice, suggesting that MT may preserve Akt2 function via inhibiting TRB3. Moreover, MT prevented tert-butyl hydroperoxide (tBHP)-reduced insulin-stimulated Akt2 phosphorylation in MT-TG cardiomyocytes, which was abolished by specific silencing of Akt2. Specific silencing of TRB3 blocked tBHP inhibition of insulin-stimulated Akt2 phosphorylation in WT cardiomyocytes, whereas overexpression of TRB3 in MT-TG cardiomyocytes and hearts abolished MT preservation of insulin-stimulated Akt2 signals and MT prevention of DCM. Most importantly, supplementation of Zn to induce MT preserved cardiac Akt2 signals and prevented DCM. These results suggest that diabetes-inhibited cardiac Akt2 function via TRB3 upregulation leads to aberrant cardiac glucose metabolism. MT preservation of cardiac Akt2 function by inhibition of TRB3 prevents DCM.
Collapse
MESH Headings
- Animals
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cells, Cultured
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/physiopathology
- Heart/drug effects
- Heart/physiopathology
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/pharmacology
- Insulin/therapeutic use
- Insulin Resistance
- Lipopolysaccharides/toxicity
- Male
- Metallothionein/genetics
- Metallothionein/metabolism
- Mice
- Mice, Mutant Strains
- Mice, Transgenic
- Myocardium/enzymology
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Organ Specificity
- Oxidants/toxicity
- Oxidative Stress/drug effects
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
Collapse
Affiliation(s)
- Junlian Gu
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Xiaoqing Yan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Xiaozhen Dai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- School of Biomedicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yuehui Wang
- Departments of Geriatrics, Cardiovascular Disorders and Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Qian Lin
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY
| | - Jian Xiao
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
| | - Shanshan Zhou
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Departments of Geriatrics, Cardiovascular Disorders and Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jian Zhang
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Departments of Geriatrics, Cardiovascular Disorders and Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Kai Wang
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Jun Zeng
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Ying Xin
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | | | - Chi Zhang
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
| | - Yang Bai
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Departments of Geriatrics, Cardiovascular Disorders and Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yan Li
- Department of Surgery, University of Louisville, Louisville, KY
| | - Paul N Epstein
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY
- Wendy L. Novak Diabetes Care Center, Louisville, KY
| | - Kupper A Wintergerst
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Wendy L. Novak Diabetes Care Center, Louisville, KY
- Division of Endocrinology, Department of Pediatrics, University of Louisville, Louisville, KY
| | - Xiaokun Li
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
| | - Yi Tan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY
- Wendy L. Novak Diabetes Care Center, Louisville, KY
| | - Lu Cai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and the First Affiliated Hospital at the Wenzhou Medical University, Wenzhou, China
- Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY
- Wendy L. Novak Diabetes Care Center, Louisville, KY
| |
Collapse
|
31
|
Zhang H, Liu X, Zhou S, Jia Y, Li Y, Song Y, Wang J, Wu H. SP600125 suppresses Keap1 expression and results in NRF2-mediated prevention of diabetic nephropathy. J Mol Endocrinol 2018; 60:145-157. [PMID: 29273684 DOI: 10.1530/jme-17-0260] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 12/22/2017] [Indexed: 12/31/2022]
Abstract
c-Jun N-terminal kinase (JNK) contributes to the pathogenesis of diabetic nephropathy (DN). The JNK inhibitor SP600125 was reported to ameliorate DN. However, the mechanism remained unclear. We previously reported that SP600125 activated nuclear factor erythroid 2-related factor 2 (NRF2), a governor of the cellular antioxidant defense system, in the aortas of the diabetic mice. Given the critical role of NRF2 in preventing DN, the present study aimed to test whether or not NRF2 is required for SP600125's protection against DN. To test the role of NRF2 in SP600125's effect, streptozotocin-induced C57BL/6 wild-type (WT) and Nrf2-knockout (KO) diabetic mice were treated in the presence or absence of SP600125, for 24 weeks. To explore the mechanism by which SP600125 activates NRF2, mouse mesangial cells (MMCs) were treated with high glucose (HG), in the presence or absence of either SP600125 or JNK siRNA. SP600125 significantly attenuated the diabetes-induced renal oxidative stress, inflammation, fibrosis, pathological change and dysfunction in the WT, but not the Nrf2 KO mice. SP600125 inactivated JNK, inhibited kelch-like ECH-associated protein 1 expression, preserved NRF2 protein and facilitated its nuclear translocation in the kidneys of the WT mice, the effects of which were similarly produced by either SP600125 or JNK siRNA in HG-treated MMCs. Further, both SP600125 and JNK siRNA alleviated HG-induced mesangial oxidative stress and expression of inflammatory and fibrotic genes. The present study demonstrates that NRF2 is required for SP600125's protection against DN. SP600125 activates NRF2 possibly via inhibition of JNK-induced Keap1 expression.
Collapse
Affiliation(s)
- Huan Zhang
- Operating TheaterChina-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiuxia Liu
- Department of Clinical LaboratoryThe Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Shanshan Zhou
- Cardiovascular CenterThe First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Ye Jia
- Department of NephrologyThe First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Ying Li
- Department of DermatologyAffiliated Hospital of Beihua University, Jilin, Jilin, People's Republic of China
| | - Yuguo Song
- Research Institute of Clinical ImmunologyAffiliated Hospital of Beihua University, Jilin, Jilin, People's Republic of China
- Research Center for Life SciencesBeihua University, Jilin, Jilin, People's Republic of China
| | - Junnan Wang
- Department of CardiologyThe Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Hao Wu
- Department of NephrologyThe Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
- The '973' National Basic Research Program of ChinaChangchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| |
Collapse
|
32
|
Gu J, Wang S, Guo H, Tan Y, Liang Y, Feng A, Liu Q, Damodaran C, Zhang Z, Keller BB, Zhang C, Cai L. Inhibition of p53 prevents diabetic cardiomyopathy by preventing early-stage apoptosis and cell senescence, reduced glycolysis, and impaired angiogenesis. Cell Death Dis 2018; 9:82. [PMID: 29362483 PMCID: PMC5833384 DOI: 10.1038/s41419-017-0093-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/01/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023]
Abstract
Elevated tumor suppressor p53 expression has been associated with heart diseases, including the diabetic heart. However, its precise role in the pathogenesis of diabetic cardiomyopathy (DCM) remains unclear. We hypothesized that the development of DCM is attributed to up-regulated p53-mediated both early cardiac cell death and persistent cell senescence, glycolytic and angiogenetic dysfunctions. The present study investigated the effect of p53 inhibition with its specific inhibitor pifithrin-α (PFT-α) on the pathogenesis of DCM and its associated mechanisms. Type 1 diabetes was induced with multiple low doses of streptozotocin. Both hyperglycemic and age-matched control mice were treated with and without PFT-α five times a week for 2 months and then sacrificed at 3 and 6 months post-diabetes. Treatment with PFT-α significantly prevented the progression of diabetes-induced cardiac remodeling and dysfunction (i.e., DCM). Mechanistically, the inhibition of p53 prevented the cardiac apoptosis during early-stage diabetes (0.5 month), attenuated diabetes-induced cell senescence (3 and 6 months), and improved both glycolytic and angiogenic defects by increasing hypoxia-induced factor (HIF)-1α protein stability and upregulating HIF-1α transcription of specific target genes at 3 and 6 months after diabetes. Therefore, the targeted inhibition of p53 in diabetic individuals may provide a novel approach for the prevention of DCM.
Collapse
Affiliation(s)
- Junlian Gu
- Ruian Center of Chinese-American Research Institute for Diabetic Complications, the Third Affiliated Hospital of the Wenzhou Medical University, Ruian, China
- Chinese-American Research Institute for Diabetic Complications, the School of Pharmaceutical Sciences of the Wenzhou Medical University, Wenzhou, China
- the Department of Pediatrics of the University of Louisville, Pediatrics Research Institute, Louisville, KY, 40202, USA
| | - Shudong Wang
- Department of Cardiology, the First Hospital of Jilin University, Changchun, 130021, China
| | - Hua Guo
- the Department of Pediatrics of the University of Louisville, Pediatrics Research Institute, Louisville, KY, 40202, USA
| | - Yi Tan
- Ruian Center of Chinese-American Research Institute for Diabetic Complications, the Third Affiliated Hospital of the Wenzhou Medical University, Ruian, China
- Chinese-American Research Institute for Diabetic Complications, the School of Pharmaceutical Sciences of the Wenzhou Medical University, Wenzhou, China
- the Department of Pediatrics of the University of Louisville, Pediatrics Research Institute, Louisville, KY, 40202, USA
| | - Yaqin Liang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Anyun Feng
- Ruian Center of Chinese-American Research Institute for Diabetic Complications, the Third Affiliated Hospital of the Wenzhou Medical University, Ruian, China
| | - Qiuju Liu
- Department of Hematology, the First Hospital of Jilin University, Changchun, 130021, China
| | - Chendil Damodaran
- Department of Urology, the University of Louisville, Louisville, KY, USA
| | - Zhiguo Zhang
- Department of Cardiology, the First Hospital of Jilin University, Changchun, 130021, China
| | - Bradley B Keller
- the Department of Pediatrics of the University of Louisville, Pediatrics Research Institute, Louisville, KY, 40202, USA
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Chi Zhang
- Ruian Center of Chinese-American Research Institute for Diabetic Complications, the Third Affiliated Hospital of the Wenzhou Medical University, Ruian, China.
- Chinese-American Research Institute for Diabetic Complications, the School of Pharmaceutical Sciences of the Wenzhou Medical University, Wenzhou, China.
| | - Lu Cai
- Ruian Center of Chinese-American Research Institute for Diabetic Complications, the Third Affiliated Hospital of the Wenzhou Medical University, Ruian, China
- Chinese-American Research Institute for Diabetic Complications, the School of Pharmaceutical Sciences of the Wenzhou Medical University, Wenzhou, China
- the Department of Pediatrics of the University of Louisville, Pediatrics Research Institute, Louisville, KY, 40202, USA
| |
Collapse
|
33
|
Jing L, Yang M, Li Y, Yu Y, Liang B, Cao L, Zhou X, Peng S, Sun Z. Metallothionein prevents doxorubicin cardiac toxicity by indirectly regulating the uncoupling proteins 2. Food Chem Toxicol 2017; 110:204-213. [PMID: 29061315 DOI: 10.1016/j.fct.2017.10.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/26/2017] [Accepted: 10/19/2017] [Indexed: 12/17/2022]
Abstract
Doxorubicin (Dox) is a broad-spectrum anticancer agent, but its clinical use is restricted due to irreversible cardiac toxicity. Metallothionein (MT) can inhibit Dox-induced cardiac toxicity. Applying a proteomics approach we determined that uncoupling proteins (UCPs) may be implicated in this process. This study was designed to examine the mechanisms of MT against Dox cardiac toxicity and the link between MT and UCP2. In vivo, wild-type (MT+/+) and MT-I/II null (MT-/-) mice were given a single dose of Dox (15 mg/kg, i.p.) and sacrificed at 4 days after Dox injection. In vitro, cardiomyocytes were prepared from MT-/- and MT+/+ neonatal mice and cardiomyocytes were pretreated with typical antioxidant NAC or the UCP2 inhibitor genipin followed by exposure to Dox. Based on the results, genipin enhanced Dox-induced oxidative injury, particularly in MT-/- cardiomyocyte. UCP2 levels in MT-/- mice were significantly lower compared to MT+/+ mice treated with Dox. Co-immunoprecipitation demonstrated that MT did not directly bind to UCP2. The NAC and Nrf2 activator oltipraz inhibit the decrease of UCP2 expression induced by Dox. Therefore, attenuating free radical damage with UCP2 help MT antagonize the Dox-induced cardiac toxicity, but does not directly bind MT. MT may regulate UCP2 expression by up-regulating Nrf2.
Collapse
Affiliation(s)
- Li Jing
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Man Yang
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Yang Li
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Yang Yu
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Baolu Liang
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Lige Cao
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Xianqing Zhou
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China
| | - Shuangqing Peng
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing 100071, PR China.
| | - Zhiwei Sun
- School of Public Health, Capital Medical University, 10 Xitoutiao, Fengtai District, Beijing 100069, PR China.
| |
Collapse
|
34
|
Yan X, Dai X, He L, Ling X, Shao M, Zhang C, Wang Y, Xiao J, Cai L, Li X, Tan Y. A Novel CXCR4 antagonist enhances angiogenesis via modifying the ischaemic tissue environment. J Cell Mol Med 2017; 21:2298-2307. [PMID: 28374486 PMCID: PMC5618675 DOI: 10.1111/jcmm.13150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/07/2017] [Indexed: 01/01/2023] Open
Abstract
Endothelial progenitor cells (EPCs) play a capital role in angiogenesis via directly participating in neo-vessel formation and secreting pro-angiogenic factors. Stromal cell-derived factor 1 (SDF-1) and its receptor CXCR4 play a critical role in the retention and quiescence of EPCs within its niche in the bone marrow. Disturbing the interaction between SDF-1 and CXCR4 is an effective strategy for EPC mobilization. We developed a novel CXCR4 antagonist P2G, a mutant protein of SDF-1β with high antagonistic activity against CXCR4 and high potency in enhancing ischaemic angiogenesis and blood perfusion. However, its direct effects on ischaemic tissue remain largely unknown. In this study, P2G was found to possess a robust capability to promote EPC infiltration and incorporation in neo-vessels, enhance the expression and function of pro-angiogenic factors, such as SDF-1, vascular endothelial growth factor and matrix metalloprotein-9, and activate cell signals involved in angiogenesis, such as proliferating cell nuclear antigen, protein kinase B (Akt), extracellular regulated protein kinases and mammalian target of rapamycin, in ischaemic tissue. Moreover, P2G can attenuate fibrotic remodelling to facilitate the recovery of ischaemic tissue. The capability of P2G in direct augmenting ischaemic environment for angiogenesis suggests that it is a potential candidate for the therapy of ischaemia diseases.
Collapse
Affiliation(s)
- Xiaoqing Yan
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of LouisvilleLouisvilleKYUSA
- Chinese‐American Pediatric Research Institute at the First Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Xiaozhen Dai
- Pediatric Research InstituteDepartment of PediatricsUniversity of LouisvilleLouisvilleKYUSA
- School of BiomedicineChengdu Medical CollegeChengduSichuanChina
| | - Luqing He
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
| | - Xiao Ling
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
| | - Minglong Shao
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
| | - Chi Zhang
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
| | - Yuehui Wang
- Department of Geriatric Medicinethe first hospital of Jilin universityChangchunJilinChina
| | - Jian Xiao
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
| | - Lu Cai
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of LouisvilleLouisvilleKYUSA
- Chinese‐American Pediatric Research Institute at the First Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Xiaokun Li
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
| | - Yi Tan
- Chinese‐American Research Institute for Diabetic ComplicationsWenzhou Medical UniversityChashan University‐townWenzhouZhejiangChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of LouisvilleLouisvilleKYUSA
- Chinese‐American Pediatric Research Institute at the First Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
35
|
The Role of Nrf2 in Cardiovascular Function and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9237263. [PMID: 29104732 PMCID: PMC5618775 DOI: 10.1155/2017/9237263] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Free radicals, reactive oxygen/nitrogen species (ROS/RNS), hydrogen sulphide, and hydrogen peroxide play an important role in both intracellular and intercellular signaling; however, their production and quenching need to be closely regulated to prevent cellular damage. An imbalance, due to exogenous sources of free radicals and chronic upregulation of endogenous production, contributes to many pathological conditions including cardiovascular disease and also more general processes involved in aging. Nuclear factor erythroid 2-like 2 (NFE2L2; commonly known as Nrf2) is a transcription factor that plays a major role in the dynamic regulation of a network of antioxidant and cytoprotective genes, through binding to and activating expression of promoters containing the antioxidant response element (ARE). Nrf2 activity is regulated by many mechanisms, suggesting that tight control is necessary for normal cell function and both hypoactivation and hyperactivation of Nrf2 are indicated in playing a role in different aspects of cardiovascular disease. Targeted activation of Nrf2 or downstream genes may prove to be a useful avenue in developing therapeutics to reduce the impact of cardiovascular disease. We will review the current status of Nrf2 and related signaling in cardiovascular disease and its relevance to current and potential treatment strategies.
Collapse
|
36
|
Wang Y, Liang B, Lau WB, Du Y, Guo R, Yan Z, Gan L, Yan W, Zhao J, Gao E, Koch W, Ma XL. Restoring diabetes-induced autophagic flux arrest in ischemic/reperfused heart by ADIPOR (adiponectin receptor) activation involves both AMPK-dependent and AMPK-independent signaling. Autophagy 2017; 13:1855-1869. [PMID: 28825851 DOI: 10.1080/15548627.2017.1358848] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Macroautophagy/autophagy is increasingly recognized as an important regulator of myocardial ischemia-reperfusion (MI-R) injury. However, whether and how diabetes may alter autophagy in response to MI-R remains unknown. Deficiency of ADIPOQ, a cardioprotective molecule, markedly increases MI-R injury. However, the role of diabetic hypoadiponectinemia in cardiac autophagy alteration after MI-R is unclear. Utilizing normal control (NC), high-fat-diet-induced diabetes, and Adipoq knockout (adipoq-/-) mice, we demonstrated that autophagosome formation was modestly inhibited and autophagosome clearance was markedly impaired in the diabetic heart subjected to MI-R. adipoq-/- largely reproduced the phenotypic alterations observed in the ischemic-reperfused diabetic heart. Treatment of diabetic and adipoq-/- mice with AdipoRon, a novel ADIPOR (adiponectin receptor) agonist, stimulated autophagosome formation, markedly increased autophagosome clearance, reduced infarct size, and improved cardiac function (P < 0.01 vs vehicle). Mechanistically, AdipoRon caused significant phosphorylation of AMPK-BECN1 (Ser93/Thr119)-class III PtdIns3K (Ser164) and enhanced lysosome protein LAMP2 expression both in vivo and in isolated adult cardiomyocytes. Pharmacological AMPK inhibition or genetic Prkaa2 mutation abolished AdipoRon-induced BECN1 (Ser93/Thr119)-PtdIns3K (Ser164) phosphorylation and AdipoRon-stimulated autophagosome formation. However, AdipoRon-induced LAMP2 expression, AdipoRon-stimulated autophagosome clearance, and AdipoRon-suppressed superoxide generation were not affected by AMPK inhibition. Treatment with MnTMPyP (a superoxide scavenger) increased LAMP2 expression and stimulated autophagosome clearance in simulated ischemic-reperfused cardiomyocytes. However, no additive effect between AdipoRon and MnTMPyP was observed. Collectively, these results demonstrate that hypoadiponectinemia impairs autophagic flux, contributing to enhanced MI-R injury in the diabetic state. ADIPOR activation restores AMPK-mediated autophagosome formation and antioxidant-mediated autophagosome clearance, representing a novel intervention effective against MI-R injury in diabetic conditions.
Collapse
Affiliation(s)
- Yajing Wang
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA.,b Center for Translational Medicine, Department of Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Bin Liang
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Wayne Bond Lau
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Yunhui Du
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Rui Guo
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Zheyi Yan
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Lu Gan
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Wenjun Yan
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Jianli Zhao
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| | - Erhe Gao
- c Center for Translational Medicine , Temple University , Philadelphia , PA , USA
| | - Walter Koch
- c Center for Translational Medicine , Temple University , Philadelphia , PA , USA
| | - Xin-Liang Ma
- a Department of Emergency Medicine , Thomas Jefferson University , Philadelphia , PA , USA.,b Center for Translational Medicine, Department of Medicine , Thomas Jefferson University , Philadelphia , PA , USA
| |
Collapse
|
37
|
HMGCS2 is a key ketogenic enzyme potentially involved in type 1 diabetes with high cardiovascular risk. Sci Rep 2017; 7:4590. [PMID: 28676675 PMCID: PMC5496911 DOI: 10.1038/s41598-017-04469-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 05/08/2017] [Indexed: 02/06/2023] Open
Abstract
Diabetes increases the risk of Cardio-vascular disease (CVD). CVD is more prevalent in type 2 diabetes (T2D) than type 1 diabetes (T1D), but the mortality risk is higher in T1D than in T2D. The pathophysiology of CVD in T1D is poorly defined. To learn more about biological pathways that are potentially involved in T1D with cardiac dysfunction, we sought to identify differentially expressed genes in the T1D heart. Our study used T1D mice with severe hyperglycemia along with significant deficits in echocardiographic measurements. Microarray analysis of heart tissue RNA revealed that the T1D mice differentially expressed 10 genes compared to control. Using Ingenuity Pathway Analysis (IPA), we showed that these genes were significantly involved in ketogenesis, cardiovascular disease, apoptosis and other toxicology functions. Of these 10 genes, the 3-Hydroxy-3-Methylglutaryl-CoA Synthase 2 (HMGCS2) was the highest upregulated gene in T1D heart. IPA analysis showed that HMGCS2 was center to many biological networks and pathways. Our data also suggested that apart from heart, the expression of HMGCS2 was also different in kidney and spleen between control and STZ treated mice. In conclusion, The HMGCS2 molecule may potentially be involved in T1D induced cardiac dysfunction.
Collapse
|
38
|
Sun W, Liu X, Zhang H, Song Y, Li T, Liu X, Liu Y, Guo L, Wang F, Yang T, Guo W, Wu J, Jin H, Wu H. Epigallocatechin gallate upregulates NRF2 to prevent diabetic nephropathy via disabling KEAP1. Free Radic Biol Med 2017; 108:840-857. [PMID: 28457936 DOI: 10.1016/j.freeradbiomed.2017.04.365] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/04/2017] [Accepted: 04/25/2017] [Indexed: 12/18/2022]
Abstract
Epigallocatechin gallate (EGCG) is the most abundant and effective green tea catechin and has been reported to attenuate diabetic nephropathy (DN). However, the mechanism by which EGCG ameliorates DN, till now, has remained unclear. EGCG is known as a potent activator of nuclear factor erythroid 2-related factor 2 (NRF2), which plays a key role in cellular defense against diabetes-induced oxidative stress and in the prevention of DN. In the present study, we tested whether NRF2 is required for EGCG protection against DN. Therefore, C57BL/6 wild type (WT) and Nrf2 knockout mice were induced to diabetes by streptozotocin, in the presence or absence of a 24-week treatment with EGCG. In the WT mice, EGCG activated Nrf2 expression and function without altering the expression of Kelch-like ECH-associated protein 1 (Keap1). Diabetes-induced renal oxidative damage, inflammation, fibrosis and albuminuria were significantly prevented by EGCG. Notably, deletion of the Nrf2 gene completely abrogated these actions of EGCG. To further determine the effect of EGCG on KEAP1/NRF2 signaling, mouse mesangial cells were treated with high glucose, in the presence of both Keap1 siRNA and EGCG. Interestingly, EGCG failed to enhance NRF2 signaling and alleviate oxidative, inflammatory and fibrotic indicators, in the presence of Keap1 siRNA. The present study demonstrated, for the first time, that NRF2 plays a critical role in EGCG protection against DN. Other findings indicated that inactivation of KEAP1 protein by EGCG may mediate EGCG function in activating NRF2.
Collapse
Affiliation(s)
- Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin St., Changchun, Jilin 130021, People's Republic of China
| | - Xiuxia Liu
- Department of Clinical Laboratory, The Second Hospital of Jilin University, 218 Ziqiang St., Changchun, Jilin 130041, People's Republic of China
| | - Haifeng Zhang
- Department of Interventional Therapy, The First Hospital of Jilin University, 71 Xinmin St., Changchun, Jilin 130021, People's Republic of China
| | - Yanyan Song
- Department of Nephrology, The Second Hospital of Jilin University, 218 Ziqiang St., Changchun, Jilin 130041, People's Republic of China
| | - Tie Li
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China; Research Institute of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China
| | - Xiaona Liu
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China; Research Institute of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China
| | - Yanze Liu
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China; Research Institute of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China
| | - Le Guo
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China; Research Institute of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China
| | - Fuchun Wang
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China; Research Institute of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China
| | - Ting Yang
- Department of Nephrology, Affiliated Hospital of Beihua University, 12 Jiefang Rd., Jilin, 132000, People's Republic of China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, 71 Xinmin St., Changchun, Jilin 130021, People's Republic of China
| | - Junduo Wu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang St., Changchun, Jilin 130041, People's Republic of China
| | - Hang Jin
- Department of Neurology, The First Hospital of Jilin University, 71 Xinmin St., Changchun, Jilin 130021, People's Republic of China.
| | - Hao Wu
- Department of Nephrology, The Second Hospital of Jilin University, 218 Ziqiang St., Changchun, Jilin 130041, People's Republic of China; Research Institute of Acupuncture and Tuina, Changchun University of Chinese Medicine, 1035 Boshuo Rd., Changchun, Jilin 130117, People's Republic of China.
| |
Collapse
|
39
|
NRF2 Plays a Critical Role in Both Self and EGCG Protection against Diabetic Testicular Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3172692. [PMID: 28698767 PMCID: PMC5494108 DOI: 10.1155/2017/3172692] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/15/2017] [Accepted: 04/06/2017] [Indexed: 01/15/2023]
Abstract
Activation of nuclear factor erythroid 2-related factor 2 (NRF2) has been found to ameliorate diabetic testicular damage (DTD) in rodents. However, it was unclear whether NRF2 is required for these approaches in DTD. Epigallocatechin gallate (EGCG) is a potent activator of NRF2 and has shown beneficial effects on multiple diabetic complications. However, the effect of EGCG has not been studied in DTD. The present study aims to explore the role of NRF2 in both self and EGCG protection against DTD. Therefore, streptozotocin-induced diabetic C57BL/6 wild type (WT) and Nrf2 knockout (KO) mice were treated in the presence or absence of EGCG, for 24 weeks. The Nrf2 KO mice exhibited more significant diabetes-induced loss in testicular weight and spermatozoa count, and increase in testicular apoptotic cell death, as compared with the WT mice. EGCG activated NRF2 expression and function, preserved testicular weight and spermatozoa count, and attenuated testicular apoptotic cell death, endoplasmic reticulum stress, inflammation, and oxidative damage in the WT diabetic mice, but not the Nrf2 KO diabetic mice. The present study demonstrated for the first time that NRF2 plays a critical role in both self and EGCG protection against DTD.
Collapse
|
40
|
Hu X, Bai T, Xu Z, Liu Q, Zheng Y, Cai L. Pathophysiological Fundamentals of Diabetic Cardiomyopathy. Compr Physiol 2017; 7:693-711. [PMID: 28333387 DOI: 10.1002/cphy.c160021] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetic cardiomyopathy (DCM) was first recognized more than four decades ago and occurred independent of cardiovascular diseases or hypertension in both type 1 and type 2 diabetic patients. The exact mechanisms underlying this disease remain incompletely understood. Several pathophysiological bases responsible for DCM have been proposed, including the presence of hyperglycemia, nonenzymatic glycosylation of large molecules (e.g., proteins), energy metabolic disturbance, mitochondrial damage and dysfunction, impaired calcium handling, reactive oxygen species formation, inflammation, cardiac cell death, and cardiac hypertrophy and fibrosis, leading to impairment of cardiac contractile functions. Increasing evidence also indicates the phenomenon called "metabolic memory" for diabetes-induced cardiovascular complications, for which epigenetic modulation seemed to play an important role, suggesting that the aforementioned pathogenic bases may be regulated by epigenetic modification. Therefore, this review aims at briefly summarizing the current understanding of the pathophysiological bases for DCM. Although how epigenetic mechanisms play a role remains incompletely understood now, extensive clinical and experimental studies have implicated its importance in regulating the cardiac responses to diabetes, which are believed to shed insight into understanding of the pathophysiological and epigenetic mechanisms for the development of DCM and its possible prevention and/or therapy. © 2017 American Physiological Society. Compr Physiol 7:693-711, 2017.
Collapse
Affiliation(s)
- Xinyue Hu
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China
- Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Tao Bai
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China
- Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Zheng Xu
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China
- Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Qiuju Liu
- Department of Hematological Disorders the First Hospital of Jilin University, Changchun, China
| | - Yang Zheng
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
- Wendy Novak Diabetes Care Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
41
|
Prevention of Streptozotocin-Induced Diabetic Nephropathy by MG132: Possible Roles of Nrf2 and I κB. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3671751. [PMID: 28373900 PMCID: PMC5360973 DOI: 10.1155/2017/3671751] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/14/2017] [Indexed: 02/07/2023]
Abstract
Our previous study showed that proteasomal inhibitor MG132 can prevent diabetic nephropathy (DN) along with upregulation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2). The present study was to investigate whether MG132 can prevent DN in wild-type and Nrf2-KO mice. Type 1 diabetes was induced in wild-type and Nrf2-KO mice by multiple low doses of streptozotocin. Two weeks after streptozotocin injection, both wild-type and Nrf2-KO mice were randomly divided into four groups: control, MG132, DM, and DM/MG132. MG132 (10 μg/kg/day) or vehicle was administered intraperitoneally for 4 months. Renal function, morphology, and biochemical changes were measured after 4-month treatment with MG132. MG132 treatment suppressed proteasomal activity in the two genotypes. In wild-type mice, MG132 attenuated diabetes-induced renal dysfunction, fibrosis, inflammation, and oxidative damage along with increased Nrf2 and IκB expression. Deletion of Nrf2 gene resulted in a partial, but significant attenuation of MG132 renal protection in Nrf2-KO mice compared with wild-type mice. MG132-increased IκB expression was not different between wild-type and Nrf2-KO mice. This work indicates that MG132 inhibits diabetes-increased proteasomal activity, resulting in Nrf2 and IκB upregulation and renal protection, which could be used as a strategy to prevent diabetic nephropathy.
Collapse
|
42
|
Wang Y, Wu H, Xin Y, Bai Y, Kong L, Tan Y, Liu F, Cai L. Sulforaphane Prevents Angiotensin II-Induced Testicular Cell Death via Activation of NRF2. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5374897. [PMID: 28191275 PMCID: PMC5278228 DOI: 10.1155/2017/5374897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/28/2016] [Accepted: 12/12/2016] [Indexed: 11/17/2022]
Abstract
Although angiotensin II (Ang II) was reported to facilitate sperm motility and intratesticular sperm transport, recent findings shed light on the efficacy of Ang II in stimulating inflammatory events in testicular peritubular cells, effect of which may play a role in male infertility. It is still unknown whether Ang II can induce testicular apoptotic cell death, which may be a more direct action of Ang II in male infertility. Therefore, the present study aims to determine whether Ang II can induce testicular apoptotic cell death and whether this action can be prevented by sulforaphane (SFN) via activating nuclear factor (erythroid-derived 2)-like 2 (NRF2), the governor of antioxidant-redox signalling. Eight-week-old male C57BL/6J wild type (WT) and Nrf2 gene knockout mice were treated with Ang II, in the presence or absence of SFN. In WT mice, SFN activated testicular NRF2 expression and function, along with a marked attenuation in Ang II-induced testicular oxidative stress, inflammation, endoplasmic reticulum stress, and apoptotic cell death. Deletion of the Nrf2 gene led to a complete abolishment of these efficacies of SFN. The present study indicated that Ang II may result in testicular apoptotic cell death, which can be prevented by SFN via the activation of NRF2.
Collapse
Affiliation(s)
- Yonggang Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, 126 Xiantai St, Changchun, Jilin 130033, China
- Pediatric Research Institute, Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Louisville, KY 40202, USA
| | - Hao Wu
- Department of Nephrology, The Second Hospital of Jilin University, 218 Ziqiang St, Changchun, Jilin 130041, China
| | - Ying Xin
- Pediatric Research Institute, Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Louisville, KY 40202, USA
- The Key Laboratory of Pathobiology, Ministry of Education, The Norman Bethune Medical College, Jilin University, Changchun, Jilin 130021, China
| | - Yang Bai
- Pediatric Research Institute, Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Louisville, KY 40202, USA
- Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin St, Changchun, Jilin 130021, China
| | - Lili Kong
- Pediatric Research Institute, Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Louisville, KY 40202, USA
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin St, Changchun, Jilin 130021, China
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Louisville, KY 40202, USA
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Feng Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China, 126 Xiantai St, Changchun, Jilin 130033, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Louisville, KY 40202, USA
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
43
|
Salem ESB, Fan GC. Pathological Effects of Exosomes in Mediating Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 998:113-138. [PMID: 28936736 DOI: 10.1007/978-981-10-4397-0_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic subjects are at risk of developing cardiovascular disease, which accounts for 60-80% of diabetes-related mortality. Atherosclerosis is still considered as a leading cause of heart failure in diabetic patients, but it could also be an intrinsic and long-term effect of contractile cardiac cells malfunction, known as diabetic cardiomyopathy (DCM). Pathologically, this cardiac dysfunction is manifested by inflammation, apoptosis, fibrosis, hypertrophy and altered cardiomyocytes metabolism. However, the underlying molecular mechanisms of DCM pathophysiology are not clearly understood. Recent and several studies have suggested that exosomes are contributed to the regulation of cell-to-cell communication. Therefore, their in-depth investigation can interpret the complex pathophysiology of DCM. Structurally, exosomes are membrane-bounded vesicles (10-200 nm in diameter), which are actively released from all types of cells and detected in all biological fluids. They carry a wide array of bioactive molecules, including mRNAs, none-coding RNAs (e.g., microRNAs, lncRNAs, circRNAs, etc), proteins and lipids. Importantly, the abundance and nature of loaded molecules inside exosomes fluctuate with cell types and pathological conditions. This chapter summarizes currently available studies on the exosomes' role in the regulation of diabetic cardiomyopathy. Specifically, the advances on the pathological effects of exosomes in diabetic cardiomyopathy as well as the therapeutic potentials and perspectives are also discussed.
Collapse
Affiliation(s)
- Esam S B Salem
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 5872 Care Mail Loc-0575, Cincinnati, OH, 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 5872 Care Mail Loc-0575, Cincinnati, OH, 45267, USA.
| |
Collapse
|
44
|
Dong W, Jia Y, Liu X, Zhang H, Li T, Huang W, Chen X, Wang F, Sun W, Wu H. Sodium butyrate activates NRF2 to ameliorate diabetic nephropathy possibly via inhibition of HDAC. J Endocrinol 2017; 232:71-83. [PMID: 27799462 DOI: 10.1530/joe-16-0322] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 10/25/2016] [Indexed: 01/04/2023]
Abstract
Oxidative stress contributes to the pathogenesis of diabetic nephropathy (DN). Nuclear factor erythroid 2-related factor 2 (NRF2) plays a key role in cellular defense against oxidative stress. NRF2 activators have shown promising preventive effects on DN. Sodium butyrate (NaB) is a known activator of NRF2. However, it is unknown whether NRF2 is required for NaB protection against DN. Therefore, streptozotocin-induced diabetic C57BL/6 Nrf2 knockout and their wild-type mice were treated in the presence or absence of NaB for 20 weeks. Diabetic mice, but not NaB-treated diabetic mice, developed significant renal oxidative damage, inflammation, apoptosis, fibrosis, pathological changes and albuminuria. NaB inhibited histone deacetylase (HDAC) activity and elevated the expression of Nrf2 and its downstream targets heme oxygenase 1 and NAD(P)H dehydrogenase quinone 1. Notably, deletion of the Nrf2 gene completely abolished NaB activation of NRF2 signaling and protection against diabetes-induced renal injury. Interestingly, the expression of Kelch-like ECH-associated protein 1, the negative regulator of NRF2, was not altered by NaB under both diabetic and non-diabetic conditions. Moreover, NRF2 nuclear translocation was not promoted by NaB. Therefore, the present study indicates, for the first time, that NRF2 plays a key role in NaB protection against DN. Other findings suggest that NaB may activate Nrf2 at the transcriptional level, possibly by the inhibition of HDAC activity.
Collapse
Affiliation(s)
- Wenpeng Dong
- Dialysis CenterDaqing Oilfield General Hospital, Daqing, Heilongjiang, People's Republic of China
- Department of NephrologyThe Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Ye Jia
- Department of NephrologyThe First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiuxia Liu
- Department of Clinical LaboratoryThe Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Huan Zhang
- Operating theatreChina-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Tie Li
- Department of Acupuncture and TuinaChangchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Wenlin Huang
- School of Science and TechnologyGeorgia Gwinnett College, Lawrenceville, Georgia, USA
| | - Xudong Chen
- Gastroenterology Department No. 1Jilin Central General Hospital, Jilin, Jilin, People's Republic of China
| | - Fuchun Wang
- Department of Acupuncture and TuinaChangchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Weixia Sun
- Department of NephrologyThe First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Hao Wu
- Department of Acupuncture and TuinaChangchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
- Department of NephrologyThe Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
45
|
Cong W, Niu C, Lv L, Ni M, Ruan D, Chi L, Wang Y, Yu Q, Zhan K, Xuan Y, Wang Y, Tan Y, Wei T, Cai L, Jin L. Metallothionein Prevents Age-Associated Cardiomyopathy via Inhibiting NF-κB Pathway Activation and Associated Nitrative Damage to 2-OGD. Antioxid Redox Signal 2016; 25:936-952. [PMID: 27477335 PMCID: PMC5144888 DOI: 10.1089/ars.2016.6648] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/20/2016] [Accepted: 07/13/2016] [Indexed: 11/12/2022]
Abstract
AIMS Cardiac-specific metallothionein (MT) overexpression extends lifespan, but the mechanism underlying the effect of MT protection against age-associated cardiovascular diseases (CVD) remains elusive. To elucidate this, male wild-type and two lines of MT-transgenic (MT-TG) mice, MM and MT-1 (cardiac-specific overexpressing MT about 10- and 80-fold, respectively) at three representative ages (2-3, 9-10, and 18-20 months), were utilized. A stable human MT2A overexpressing cardiomyocytes (H9c2MT7) was also introduced. RESULTS Histomorphology and echocardiographic analysis revealed that age-associated cardiac hypertrophy, remodeling, and dysfunction were ameliorated in MT-TG mice. Also, aging-accompanied NF-κB activation, characterized by increased nuclear p65 translocation, elevated DNA-binding activity, and upregulation of inflammatory cytokines, was largely attenuated by MT overexpression. Treatment of H9c2 cardiomyocytes with tumor necrosis factor-α (TNF-α), which mimicked an inflammatory environment, significantly increased NF-κB activity, and some age-related phenotypes appeared. The NF-κB activation was further proved to be pivotal for both age-associated and TNF-α-induced nitrative damage to cardiac 2-oxoglutarate dehydrogenase (2-OGD) by virtue of NF-κB p65 gene silencing. MT inhibited NF-κB activation and associated nitrative damage to cardiac 2-OGD in both old MT-TG hearts and TNF-α-treated H9c2MT7 cardiomyocytes; these protective effects were abolished in H9c2MT7 cardiomyocytes by MT-specific gene silencing. Innovation and Conclusion: Together, these findings indicate that the protective effects of MT against age-associated CVD can be attributed mainly to its role in NF-κB inhibition and resultant alleviation of nitrative damage to 2-OGD. Antioxid. Redox Signal. 25, 936-952.
Collapse
Affiliation(s)
- Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Chao Niu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Lingchun Lv
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
| | - Maowei Ni
- Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | - Dandan Ruan
- The Health Examination Center, the 117th Hospital of Chinese People's Liberation Army, Hangzhou, P.R. China
| | - Lisha Chi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yang Wang
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, P.R. China
| | - Qing Yu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Kungao Zhan
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Yuanhu Xuan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yuehui Wang
- The First Hospital of Jilin University, Changchun, P.R. China
| | - Yi Tan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville, Louisville, Kentucky
| | - Tiemin Wei
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
| | - Lu Cai
- The First Hospital of Jilin University, Changchun, P.R. China
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville, Louisville, Kentucky
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| |
Collapse
|
46
|
Karri VVSR, Kuppusamy G, Talluri SV, Mannemala SS, Kollipara R, Wadhwani AD, Mulukutla S, Raju KRS, Malayandi R. Curcumin loaded chitosan nanoparticles impregnated into collagen-alginate scaffolds for diabetic wound healing. Int J Biol Macromol 2016; 93:1519-1529. [DOI: 10.1016/j.ijbiomac.2016.05.038] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022]
|
47
|
Verma SK, Garikipati VNS, Kishore R. Mitochondrial dysfunction and its impact on diabetic heart. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1098-1105. [PMID: 27593695 DOI: 10.1016/j.bbadis.2016.08.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction and associated oxidative stress are strongly linked to cardiovascular, neurodegenerative, and age associated disorders. More specifically cardiovascular diseases are common in patients with diabetes and significant contributor to the high mortality rates associated with diabetes. Studies have shown that the heart failure risk is increased in diabetic patients even after adjusting for coronary artery disease and hypertension. Although the actual basis of the increased heart failure risk is multifactorial, increasing evidences suggest that imbalances in mitochondrial function and associated oxidative stress play an important role in this process. This review summarizes these abnormalities in mitochondrial function and discusses potential underlying mechanisms. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Suresh Kumar Verma
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| | | | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
48
|
Wu H, Kong L, Tan Y, Epstein PN, Zeng J, Gu J, Liang G, Kong M, Chen X, Miao L, Cai L. C66 ameliorates diabetic nephropathy in mice by both upregulating NRF2 function via increase in miR-200a and inhibiting miR-21. Diabetologia 2016; 59:1558-1568. [PMID: 27115417 PMCID: PMC5260795 DOI: 10.1007/s00125-016-3958-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/23/2016] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Diabetic nephropathy is the leading cause of end-stage renal disease. Previously we reported that C66, a novel analogue of curcumin with a very high bioavailability, ameliorated diabetic nephropathy in mice, with little known about the mechanism. The present study aimed to define the mechanism by which C66 ameliorates diabetic nephropathy. METHODS Our aim was to discover whether C66 acts through the activation of nuclear factor (erythroid-derived 2)-like 2 (NFE2L2 or NRF2), which governs the antioxidant response. Streptozotocin-induced Nrf2 (also known as Nfe2l2)-knockout and wild-type (WT) diabetic mice were treated with C66. To determine whether the actions of C66 on NRF2 are mediated by microRNA (miR)-200a, WT diabetic mice were treated with C66 in the presence or absence of an in vivo miR-200a inhibitor (locked nucleic acid-modified anti-miR-200a [LNA-200a]) for 6 months. To determine whether miR-21 downregulation provided an NRF2-independent basis for C66 protection, Nrf2-knockout diabetic mice were treated with either C66 or an inhibitor of miR-21 (locked nucleic acid-modified anti-miR-21 [LNA-21]). RESULTS Deletion of Nrf2 partially abolished diabetic nephropathy protection by C66, confirming the requirement of NRF2 for this protection. Diabetic mice, but not C66-treated diabetic mice, developed significant albuminuria, renal oxidative damage and fibrosis. C66 upregulated renal miR-200a, inhibited kelch-like ECH-associated protein 1 and induced NRF2 function, effects that were prevented by LNA-200a. However, LNA-200a only partially reduced the protection afforded by C66, suggesting the existence of miR-200a/NRF2-independent mechanisms for C66 protection. C66 was also found to inhibit diabetes induction of miR-21. Both C66 and LNA-21 produced similar reductions in miR-21, albuminuria and renal fibrosis. CONCLUSIONS/INTERPRETATION The present study indicates that in addition to upregulating NRF2 by increasing miR-200a, C66 also protects against diabetic nephropathy by inhibiting miR-21.
Collapse
Affiliation(s)
- Hao Wu
- Department of Nephrology, the Second Hospital of Jilin University, 218 Ziqiang St, Changchun, Jilin, 130041, People's Republic of China
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Lili Kong
- Department of Nephrology, the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Yi Tan
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Paul N Epstein
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Jun Zeng
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Junlian Gu
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lining Miao
- Department of Nephrology, the Second Hospital of Jilin University, 218 Ziqiang St, Changchun, Jilin, 130041, People's Republic of China.
| | - Lu Cai
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA.
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
49
|
Myers RB, Fomovsky GM, Lee S, Tan M, Wang BF, Patwari P, Yoshioka J. Deletion of thioredoxin-interacting protein improves cardiac inotropic reserve in the streptozotocin-induced diabetic heart. Am J Physiol Heart Circ Physiol 2016; 310:H1748-59. [PMID: 27037370 DOI: 10.1152/ajpheart.00051.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/25/2016] [Indexed: 02/05/2023]
Abstract
Although the precise pathogenesis of diabetic cardiac damage remains unclear, potential mechanisms include increased oxidative stress, autonomic nervous dysfunction, and altered cardiac metabolism. Thioredoxin-interacting protein (Txnip) was initially identified as an inhibitor of the antioxidant thioredoxin but is now recognized as a member of the arrestin superfamily of adaptor proteins that classically regulate G protein-coupled receptor signaling. Here we show that Txnip plays a key role in diabetic cardiomyopathy. High glucose levels induced Txnip expression in rat cardiomyocytes in vitro and in the myocardium of streptozotocin-induced diabetic mice in vivo. While hyperglycemia did not induce cardiac dysfunction at baseline, β-adrenergic challenge revealed a blunted myocardial inotropic response in diabetic animals (24-wk-old male and female C57BL/6;129Sv mice). Interestingly, diabetic mice with cardiomyocyte-specific deletion of Txnip retained a greater cardiac response to β-adrenergic stimulation than wild-type mice. This benefit in Txnip-knockout hearts was not related to the level of thioredoxin activity or oxidative stress. Unlike the β-arrestins, Txnip did not interact with β-adrenergic receptors to desensitize downstream signaling. However, our proteomic and functional analyses demonstrated that Txnip inhibits glucose transport through direct binding to glucose transporter 1 (GLUT1). An ex vivo analysis of perfused hearts further demonstrated that the enhanced functional reserve afforded by deletion of Txnip was associated with myocardial glucose utilization during β-adrenergic stimulation. These data provide novel evidence that hyperglycemia-induced Txnip is responsible for impaired cardiac inotropic reserve by direct regulation of insulin-independent glucose uptake through GLUT1 and plays a role in the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ronald B Myers
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gregory M Fomovsky
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Samuel Lee
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Max Tan
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Bing F Wang
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Parth Patwari
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jun Yoshioka
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Zhang J, Cheng Y, Gu J, Wang S, Zhou S, Wang Y, Tan Y, Feng W, Fu Y, Mellen N, Cheng R, Ma J, Zhang C, Li Z, Cai L. Fenofibrate increases cardiac autophagy via FGF21/SIRT1 and prevents fibrosis and inflammation in the hearts of Type 1 diabetic mice. Clin Sci (Lond) 2016; 130:625-641. [PMID: 26795437 DOI: 10.1042/cs20150623] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/21/2016] [Indexed: 02/02/2023]
Abstract
Fenofibrate (FF), as a peroxisome-proliferator-activated receptor α (PPARα) agonist, has been used clinically for decades to lower lipid levels. In the present study, we examined whether FF can be repurposed to prevent the pathogenesi of the heart in Type 1 diabetes and to describe the underlying mechanism of its action. Streptozotocin (STZ)-induced diabetic mice and their age-matched control mice were treated with vehicle or FF by gavage every other day for 3 or 6 months. FF prevented diabetes-induced cardiac dysfunction (e.g. decreased ejection fraction and hypertrophy), inflammation and remodelling. FF also increased cardiac expression of fibroblast growth factor 21 (FGF21) and sirtuin 1 (Sirt1) in non-diabetic and diabetic conditions. Deletion of FGF21 gene (FGF21-KO) worsened diabetes-induced pathogenic effects in the heart. FF treatment prevented heart deterioration in the wild-type diabetic mice, but could not do so in the FGF21-KO diabetic mice although the systemic lipid profile was lowered in both wild-type and FGF21-KO diabetic mice. Mechanistically, FF treatment prevented diabetes-impaired autophagy, reflected by increased microtubule-associated protein 1A/1B-light chain 3, in the wild-type diabetic mice but not in the FGF21-KO diabetic mice. Studies with H9C2 cells in vitro demonstrated that exposure to high glucose (HG) significantly increased inflammatory response, oxidative stress and pro-fibrotic response and also significantly inhibited autophagy. These effects of HG were prevented by FF treatment. Inhibition of either autophagy by 3-methyladenine (3MA) or Sirt1 by sirtinol (SI) abolished FF's prevention of HG-induced effects. These results suggested that FF could prevent Type 1 diabetes-induced pathological and functional abnormalities of the heart by increasing FGF21 that may up-regulate Sirt1-mediated autophagy.
Collapse
MESH Headings
- Animals
- Autophagy/drug effects
- Blood Glucose/metabolism
- Cell Line
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/pathology
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/prevention & control
- Fenofibrate/pharmacology
- Fibroblast Growth Factors/deficiency
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Fibrosis
- Histone Deacetylase Inhibitors/pharmacology
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/prevention & control
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocarditis/enzymology
- Myocarditis/etiology
- Myocarditis/pathology
- Myocarditis/physiopathology
- Myocarditis/prevention & control
- Myocardium/enzymology
- Myocardium/pathology
- Oxidative Stress/drug effects
- Rats
- Signal Transduction/drug effects
- Sirtuin 1/metabolism
- Time Factors
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People's Hospital of Liaoning Province, Shenyang 110016, China The Chinese-American Research Institute for Diabetic Complications, the Wenzhou Medical University, Wenzhou 325035, China Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A
| | - Yanli Cheng
- Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A. The First Hospital of Jilin University, Changchun 130021, China
| | - Junlian Gu
- Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A
| | - Shudong Wang
- Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A. The First Hospital of Jilin University, Changchun 130021, China
| | - Shanshan Zhou
- Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A. The First Hospital of Jilin University, Changchun 130021, China
| | - Yuehui Wang
- The First Hospital of Jilin University, Changchun 130021, China
| | - Yi Tan
- The Chinese-American Research Institute for Diabetic Complications, the Wenzhou Medical University, Wenzhou 325035, China Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A. Departments of Medicine and Pharmacology & Toxicology, University of Louisville, Louisville 40202, KY, U.S.A
| | - Wenke Feng
- Departments of Medicine and Pharmacology & Toxicology, University of Louisville, Louisville 40202, KY, U.S.A
| | - Yaowen Fu
- The First Hospital of Jilin University, Changchun 130021, China
| | - Nicholas Mellen
- Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A
| | - Rui Cheng
- Department of Physiology, the University of Oklahoma Health Sciences Center, Oklahoma City 73104, OK, U.S.A
| | - Jianxing Ma
- Department of Physiology, the University of Oklahoma Health Sciences Center, Oklahoma City 73104, OK, U.S.A
| | - Chi Zhang
- The Chinese-American Research Institute for Diabetic Complications, the Wenzhou Medical University, Wenzhou 325035, China
| | - Zhanquan Li
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People's Hospital of Liaoning Province, Shenyang 110016, China
| | - Lu Cai
- The Chinese-American Research Institute for Diabetic Complications, the Wenzhou Medical University, Wenzhou 325035, China Kosair Children Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville 40202, KY, U.S.A. Departments of Medicine and Pharmacology & Toxicology, University of Louisville, Louisville 40202, KY, U.S.A.
| |
Collapse
|