1
|
Du L, Wang X, Guo Y, Tao T, Wu H, Xu X, Zhang B, Chen T, Xu Q, Guo X. Altered lipid metabolism promoting cardiac fibrosis is mediated by CD34 + cell-derived FABP4 + fibroblasts. Exp Mol Med 2024; 56:1869-1886. [PMID: 39198543 PMCID: PMC11372182 DOI: 10.1038/s12276-024-01309-9] [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: 10/09/2023] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 09/01/2024] Open
Abstract
Hyperlipidemia and hypertension might play a role in cardiac fibrosis, in which a heterogeneous population of fibroblasts seems important. However, it is unknown whether CD34+ progenitor cells are involved in the pathogenesis of heart fibrosis. This study aimed to explore the mechanism of CD34+ cell differentiation in cardiac fibrosis during hyperlipidemia. Through the analysis of transcriptomes from 50,870 single cells extracted from mouse hearts and 76,851 single cells from human hearts, we have effectively demonstrated the evolving cellular landscape throughout cardiac fibrosis. Disturbances in lipid metabolism can accelerate the development of fibrosis. Through the integration of bone marrow transplantation models and lineage tracing, our study showed that hyperlipidemia can expedite the differentiation of non-bone marrow-derived CD34+ cells into fibroblasts, particularly FABP4+ fibroblasts, in response to angiotensin II. Interestingly, the partial depletion of CD34+ cells led to a notable reduction in triglycerides in the heart, mitigated fibrosis, and improved cardiac function. Furthermore, immunostaining of human heart tissue revealed colocalization of CD34+ cells and fibroblasts. Mechanistically, our investigation of single-cell RNA sequencing data through pseudotime analysis combined with in vitro cellular studies revealed the crucial role of the PPARγ/Akt/Gsk3β pathway in orchestrating the differentiation of CD34+ cells into FABP4+ fibroblasts. Through our study, we generated valuable insights into the cellular landscape of CD34+ cell-derived cells in the hypertrophic heart with hyperlipidemia, indicating that the differentiation of non-bone marrow-derived CD34+ cells into FABP4+ fibroblasts during this process accelerates lipid accumulation and promotes heart failure via the PPARγ/Akt/Gsk3β pathway.
Collapse
Affiliation(s)
- Luping Du
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Tao
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Wu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Health care, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Liao Y, Zhang Z, Ouyang L, Mi B, Liu G. Engineered Extracellular Vesicles in Wound Healing: Design, Paradigms, and Clinical Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307058. [PMID: 37806763 DOI: 10.1002/smll.202307058] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/20/2023] [Indexed: 10/10/2023]
Abstract
The severe quality of life and economic burden imposed by non-healing skin wounds, infection risks, and treatment costs are affecting millions of patients worldwide. To mitigate these challenges, scientists are relentlessly seeking effective treatment measures. In recent years, extracellular vesicles (EVs) have emerged as a promising cell-free therapy strategy, attracting extensive attention from researchers. EVs mediate intercellular communication, possessing excellent biocompatibility and stability. These features make EVs a potential tool for treating a plethora of diseases, including those related to wound repair. However, there is a growing focus on the engineering of EVs to overcome inherent limitations such as low production, relatively fixed content, and targeting capabilities of natural EVs. This engineering could improve both the effectiveness and specificity of EVs in wound repair treatments. In light of this, the present review will introduce the latest progress in the design methods and experimental paradigms of engineered EVs applied in wound repair. Furthermore, it will comprehensively analyze the current clinical research status and prospects of engineered EVs within this field.
Collapse
Affiliation(s)
- Yuheng Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zhenhe Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Lizhi Ouyang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| |
Collapse
|
3
|
Cummins ML, Delmonte G, Wechsler S, Schlesinger JJ. Alleviating mitochondrial dysfunction in diabetic cardiomyopathy through the Adipsin and Irak2 pathways. Mil Med Res 2024; 11:11. [PMID: 38303084 PMCID: PMC10832134 DOI: 10.1186/s40779-024-00513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Affiliation(s)
- Mabel L Cummins
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA.
| | - Grace Delmonte
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Skylar Wechsler
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Joseph J Schlesinger
- Division of Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| |
Collapse
|
4
|
O’Brien JG, Willis AB, Long AM, Kwon J, Lee G, Li FW, Page PG, Vo AH, Hadhazy M, Spencer MJ, Crosbie RH, Demonbreun AR, McNally EM. The super-healing MRL strain promotes muscle growth in muscular dystrophy through a regenerative extracellular matrix. JCI Insight 2024; 9:e173246. [PMID: 38175727 PMCID: PMC11143963 DOI: 10.1172/jci.insight.173246] [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: 06/21/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
The Murphy Roths Large (MRL) mouse strain has "super-healing" properties that enhance recovery from injury. In mice, the DBA/2J strain intensifies many aspects of muscular dystrophy, so we evaluated the ability of the MRL strain to suppress muscular dystrophy in the Sgcg-null mouse model of limb girdle muscular dystrophy. A comparative analysis of Sgcg-null mice in the DBA/2J versus MRL strains showed greater myofiber regeneration, with reduced structural degradation of muscle in the MRL strain. Transcriptomic profiling of dystrophic muscle indicated strain-dependent expression of extracellular matrix (ECM) and TGF-β signaling genes. To investigate the MRL ECM, cellular components were removed from dystrophic muscle sections to generate decellularized myoscaffolds. Decellularized myoscaffolds from dystrophic mice in the protective MRL strain had significantly less deposition of collagen and matrix-bound TGF-β1 and TGF-β3 throughout the matrix. Dystrophic myoscaffolds from the MRL background, but not the DBA/2J background, were enriched in myokines like IGF-1 and IL-6. C2C12 myoblasts seeded onto decellularized matrices from Sgcg-/- MRL and Sgcg-/- DBA/2J muscles showed the MRL background induced greater myoblast differentiation compared with dystrophic DBA/2J myoscaffolds. Thus, the MRL background imparts its effect through a highly regenerative ECM, which is active even in muscular dystrophy.
Collapse
Affiliation(s)
- Joseph G. O’Brien
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alexander B. Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ashlee M. Long
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason Kwon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - GaHyun Lee
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Frank W. Li
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Patrick G.T. Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andy H. Vo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Rachelle H. Crosbie
- Department of Integrative Biology and Physiology, Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
5
|
Jiang MY, Man WR, Zhang XB, Zhang XH, Duan Y, Lin J, Zhang Y, Cao Y, Wu DX, Shu XF, Xin L, Wang H, Zhang X, Li CY, Gu XM, Zhang X, Sun DD. Adipsin inhibits Irak2 mitochondrial translocation and improves fatty acid β-oxidation to alleviate diabetic cardiomyopathy. Mil Med Res 2023; 10:63. [PMID: 38072993 PMCID: PMC10712050 DOI: 10.1186/s40779-023-00493-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) causes the myocardium to rely on fatty acid β-oxidation for energy. The accumulation of intracellular lipids and fatty acids in the myocardium usually results in lipotoxicity, which impairs myocardial function. Adipsin may play an important protective role in the pathogenesis of DCM. The aim of this study is to investigate the regulatory effect of Adipsin on DCM lipotoxicity and its molecular mechanism. METHODS A high-fat diet (HFD)-induced type 2 diabetes mellitus model was constructed in mice with adipose tissue-specific overexpression of Adipsin (Adipsin-Tg). Liquid chromatography-tandem mass spectrometry (LC-MS/MS), glutathione-S-transferase (GST) pull-down technique, Co-immunoprecipitation (Co-IP) and immunofluorescence colocalization analyses were used to investigate the molecules which can directly interact with Adipsin. The immunocolloidal gold method was also used to detect the interaction between Adipsin and its downstream modulator. RESULTS The expression of Adipsin was significantly downregulated in the HFD-induced DCM model (P < 0.05). Adipose tissue-specific overexpression of Adipsin significantly improved cardiac function and alleviated cardiac remodeling in DCM (P < 0.05). Adipsin overexpression also alleviated mitochondrial oxidative phosphorylation function in diabetic stress (P < 0.05). LC-MS/MS analysis, GST pull-down technique and Co-IP studies revealed that interleukin-1 receptor-associated kinase-like 2 (Irak2) was a downstream regulator of Adipsin. Immunofluorescence analysis also revealed that Adipsin was co-localized with Irak2 in cardiomyocytes. Immunocolloidal gold electron microscopy and Western blotting analysis indicated that Adipsin inhibited the mitochondrial translocation of Irak2 in DCM, thus dampening the interaction between Irak2 and prohibitin (Phb)-optic atrophy protein 1 (Opa1) on mitochondria and improving the structural integrity and function of mitochondria (P < 0.05). Interestingly, in the presence of Irak2 knockdown, Adipsin overexpression did not further alleviate myocardial mitochondrial destruction and cardiac dysfunction, suggesting a downstream role of Irak2 in Adipsin-induced responses (P < 0.05). Consistent with these findings, overexpression of Adipsin after Irak2 knockdown did not further reduce the accumulation of lipids and their metabolites in the cardiac myocardium, nor did it enhance the oxidation capacity of cardiomyocytes expose to palmitate (PA) (P < 0.05). These results indicated that Irak2 may be a downstream regulator of Adipsin. CONCLUSIONS Adipsin improves fatty acid β-oxidation and alleviates mitochondrial injury in DCM. The mechanism is related to Irak2 interaction and inhibition of Irak2 mitochondrial translocation.
Collapse
Affiliation(s)
- Meng-Yuan Jiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Wan-Rong Man
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xue-Bin Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Hua Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yang Cao
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - De-Xi Wu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Fei Shu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Lei Xin
- Department of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Hao Wang
- Department of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Cong-Ye Li
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Ming Gu
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, 710032, China
| | - Xuan Zhang
- Institute for Hospital Management Research, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Dong-Dong Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
O’Brien JG, Willis AB, Long AM, Kwon J, Lee G, Li F, Page PG, Vo AH, Hadhazy M, Crosbie RH, Demonbreun AR, McNally EM. The super-healing MRL strain promotes muscle growth in muscular dystrophy through a regenerative extracellular matrix. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547098. [PMID: 37425960 PMCID: PMC10327155 DOI: 10.1101/2023.06.29.547098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Genetic background shifts the severity of muscular dystrophy. In mice, the DBA/2J strain confers a more severe muscular dystrophy phenotype, whereas the Murphy's Roth Large (MRL) strain has "super-healing" properties that reduce fibrosis. A comparative analysis of the Sgcg null model of Limb Girdle Muscular Dystrophy in the DBA/2J versus MRL strain showed the MRL background was associated with greater myofiber regeneration and reduced structural degradation of muscle. Transcriptomic profiling of dystrophic muscle in the DBA/2J and MRL strains indicated strain-dependent expression of the extracellular matrix (ECM) and TGF-β signaling genes. To investigate the MRL ECM, cellular components were removed from dystrophic muscle sections to generate decellularized "myoscaffolds". Decellularized myoscaffolds from dystrophic mice in the protective MRL strain had significantly less deposition of collagen and matrix-bound TGF-β1 and TGF-β3 throughout the matrix, and dystrophic myoscaffolds from the MRL background were enriched in myokines. C2C12 myoblasts were seeded onto decellularized matrices from Sgcg-/- MRL and Sgcg-/- DBA/2J matrices. Acellular myoscaffolds from the dystrophic MRL background induced myoblast differentiation and growth compared to dystrophic myoscaffolds from the DBA/2J matrices. These studies establish that the MRL background also generates its effect through a highly regenerative ECM, which is active even in muscular dystrophy.
Collapse
Affiliation(s)
- Joseph G. O’Brien
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alexander B. Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ashlee M. Long
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jason Kwon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - GaHyun Lee
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Frank Li
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Patrick G.T. Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rachelle H. Crosbie
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA; Department of Neurology David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Tian H, Zhao X, Zhang Y, Xia Z. Abnormalities of glucose and lipid metabolism in myocardial ischemia-reperfusion injury. Biomed Pharmacother 2023; 163:114827. [PMID: 37141734 DOI: 10.1016/j.biopha.2023.114827] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/23/2023] [Accepted: 04/30/2023] [Indexed: 05/06/2023] Open
Abstract
Myocardial ischemia-reperfusion injury is a common condition in cardiovascular diseases, and the mechanism of its occurrence involves multiple complex metabolic pathways and signaling pathways. Among these pathways, glucose metabolism and lipid metabolism play important roles in regulating myocardial energy metabolism. Therefore, this article focuses on the roles of glucose metabolism and lipid metabolism in myocardial ischemia-reperfusion injury, including glycolysis, glucose uptake and transport, glycogen metabolism and the pentose phosphate pathway; and triglyceride metabolism, fatty acid uptake and transport, phospholipid metabolism, lipoprotein metabolism, and cholesterol metabolism. Finally, due to the different alterations and development of glucose metabolism and lipid metabolism in myocardial ischemia-reperfusion, there are also complex interregulatory relationships between them. In the future, modulating the equilibrium between glucose metabolism and lipid metabolism in cardiomyocytes and ameliorating aberrations in myocardial energy metabolism represent highly promising novel strategies for addressing myocardial ischemia-reperfusion injury. Therefore, a comprehensive exploration of glycolipid metabolism can offer novel theoretical and clinical insights into the prevention and treatment of myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hao Tian
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xiaoshuai Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yuxi Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
8
|
Ding N, Zheng C. Secreted frizzled-related protein 5 promotes angiogenesis of human umbilical vein endothelial cells and alleviates myocardial injury in diabetic mice with myocardial infarction by inhibiting Wnt5a/JNK signaling. Bioengineered 2022; 13:11656-11667. [PMID: 35506262 PMCID: PMC9275896 DOI: 10.1080/21655979.2022.2070964] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study is to investigate whether secreted frizzled-related protein 5 (SFRP5) affects the proliferation, migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) induced by high glucose (HG). HUVECs were treated with different concentrations of glucose. MTT, wound healing, angiogenesis, and ELISA assays were used to detect cell cytotoxicity, migration, tube formation, and VEGF165 and VEGF165b levels, respectively. The mice model of type 2 diabetes mellitus (T2DM) complicated with myocardial infarction (MI) was established. SFRP5 was injected intrabitoneally for 2 weeks. cardiac output (CO), left ventricular ejection fraction (LVEF) and left ventricular shortening fraction (LVSF) were detected by echocardiography. Western blot was used to detect the protein levels of SFRP5, Wnt5a, JNK1/2/3, p-JNK1/2/3, TGF-β1, Caspase3, Bax, and Bcl-2. The expression of SFRP5 was declined in HG-induced HUVECs and T2DM-MI. Intervention of SFRP5 promoted the migration of HUVECs and angiogenesis, as evidenced by a lower expression of Bax and caspase3, but a higher expression of Bcl-2. Meanwhile, SFRP5 inhibition repress Wnt5a and p-JNK expression. Howerver, The JNK inhibitor (SP600125) enhanced the down-regulation of Wnt5a/JNK pathway proteins by SFRP5. SFRP5 intervention increased the levels of CO, LVSF, and LVEF in T2DM-MI mice. SFRP5 inhibited myocardial pathological injury and fibrosis in T2DM-MI mice and SFRP5 could down-regulate Wnt5a and p-JNK1/2/3 activation. SFRP5 promotes the proliferation, migration and angiogenesis of HUVECs induced by HG, and inhibits cardiac dysfunction, pathological damage, fibrosis, and myocardial angiogenesis in diabetic myocardial ischemia mice, which is achieved by inhibiting Wnt5a/JNK signaling.
Collapse
Affiliation(s)
- Nian Ding
- Clinical College of Traditional Chinese medicine, Hubei University of Chinese Medicine, Wuhan, China.,Medical Ward, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Chenghong Zheng
- Clinical College of Traditional Chinese medicine, Hubei University of Chinese Medicine, Wuhan, China.,Medical Ward, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
9
|
Kwiatkowski G, Bar A, Jasztal A, Chłopicki S. MRI-based in vivo detection of coronary microvascular dysfunction before alterations in cardiac function induced by short-term high-fat diet in mice. Sci Rep 2021; 11:18915. [PMID: 34556779 PMCID: PMC8460671 DOI: 10.1038/s41598-021-98401-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelial dysfunction is one of the hallmarks of vascular abnormalities in metabolic diseases and has been repeatedly demonstrated in coronary and peripheral circulation in mice fed high-fat diet (HFD), particularly after long-term HFD. However, the temporal relationship between development of coronary microvascular endothelial dysfunction and deterioration in diastolic and systolic cardiac function after short-term feeding with HFD has not yet been studied. This study aimed to correlate the changes in coronary microvascular endothelial function and global cardiac performance indices in vivo after short-term feeding with HFD in mice. Short-term feeding with a HFD (60% fat + 1% cholesterol) resulted in severely impaired coronary microvascular function, as evidenced by the diminished effect of nitric oxide synthase inhibition (by L-NAME) assessed using T1 mapping via in vivo MRI. Deterioration of coronary microvascular function was detected as early as after 7 days of HFD and further declined after 8 weeks on a HFD. HFD-induced coronary microvascular dysfunction was not associated with impaired myocardial capillary density and was present before systemic insulin resistance assessed by a glucose tolerance test. Basal coronary flow and coronary reserve, as assessed using the A2A adenosine receptor agonist regadenoson, were also not altered in HFD-fed mice. Histological analysis did not reveal cardiomyocyte hypertrophy or fibrosis. Increased lipid accumulation in cardiomyocytes was detected as early as after 7 days of HFD and remained at a similar level at 8 weeks on a HFD. Multiparametric cardiac MRI revealed a reduction in systolic heart function, including decreased ejection rate, increased end-systolic volume and decreased myocardial strain in diastole with impaired ejection fraction, but not until 4 weeks of HFD. Short-term feeding with HFD resulted in early endothelial dysfunction in coronary microcirculation that preceded alteration in cardiac function and systemic insulin resistance.
Collapse
Affiliation(s)
- Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Stefan Chłopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, ul. Bobrzynskiego 14, 30-348, Kraków, Poland.
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Kraków, Poland.
| |
Collapse
|
10
|
Wingard MC, Dalal S, Shook PL, Myers R, Connelly BA, Thewke DP, Singh M, Singh K. Deficiency of ataxia-telangiectasia mutated kinase modulates functional and biochemical parameters of the heart in response to Western-type diet. Am J Physiol Heart Circ Physiol 2021; 320:H2324-H2338. [PMID: 33929897 PMCID: PMC8289354 DOI: 10.1152/ajpheart.00990.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/31/2021] [Accepted: 04/14/2021] [Indexed: 02/08/2023]
Abstract
Ataxia-telangiectasia mutated (ATM) kinase deficiency exacerbates heart dysfunction late after myocardial infarction. Here, we hypothesized that ATM deficiency modulates Western-type diet (WD)-induced cardiac remodeling with an emphasis on functional and biochemical parameters of the heart. Weight gain was assessed in male wild-type (WT) and ATM heterozygous knockout (hKO) mice on weekly basis, whereas cardiac functional and biochemical parameters were measured 14 wk post-WD. hKO-WD mice exhibited rapid body weight gain at weeks 5, 6, 7, 8, and 10 versus WT-WD. WD decreased percent fractional shortening and ejection fraction, and increased end-systolic volumes and diameters to a similar extent in both genotypes. However, WD decreased stroke volume, cardiac output, peak velocity of early ventricular filling, and aortic ejection time and increased isovolumetric relaxation time (IVRT) and Tei index versus WT-NC (normal chow). Conversely, IVRT, isovolumetric contraction time, and Tei index were lower in hKO-WD versus hKO-NC and WT-WD. Myocyte apoptosis and hypertrophy were higher in hKO-WD versus WT-WD. WD increased fibrosis and expression of collagen-1α1, matrix metalloproteinase (MMP)-2, and MMP-9 in WT. WD enhanced AMPK activation, while decreasing mTOR activation in hKO. Akt and IKK-α/β activation, and Bax, PARP-1, and Glut-4 expression were higher in WT-WD versus WT-NC, whereas NF-κB activation and Glut-4 expression were lower in hKO-WD versus hKO-NC. Circulating concentrations of IL-12(p70), eotaxin, IFN-γ, macrophage inflammatory protein (MIP)-1α, and MIP-1β were higher in hKO-WD versus WT-WD. Thus, ATM deficiency accelerates weight gain, induces systolic dysfunction with increased preload, and associates with increased apoptosis, hypertrophy, and inflammation in response to WD.NEW & NOTEWORTHY Ataxia-telangiectasia mutated (ATM) kinase deficiency in humans associates with enhanced susceptibility to ischemic heart disease. Here, we provide evidence that ATM deficiency accelerates body weight gain and associates with increased cardiac preload, hypertrophy, and apoptosis in mice fed with Western-type diet (WD). Further investigations of the role of ATM deficiency in WD-induced alterations in function and biochemical parameters of the heart may provide clinically applicable information on treatment and/or nutritional counseling for patients with ATM deficiency.
Collapse
Affiliation(s)
- Mary C Wingard
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Suman Dalal
- Department of Health Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Paige L Shook
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Rachel Myers
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Barbara A Connelly
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- James H Quillen Veterans Affairs Medical Center, East Tennessee State University, Johnson City, Tennessee
| | - Douglas P Thewke
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Mahipal Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Krishna Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
- James H Quillen Veterans Affairs Medical Center, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
11
|
Alsheikh HAM, Metge BJ, Ha CM, Hinshaw DC, Mota MSV, Kammerud SC, Lama-Sherpa T, Sharafeldin N, Wende AR, Samant RS, Shevde LA. Normalizing glucose levels reconfigures the mammary tumor immune and metabolic microenvironment and decreases metastatic seeding. Cancer Lett 2021; 517:24-34. [PMID: 34052331 DOI: 10.1016/j.canlet.2021.05.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/09/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
Obesity and diabetes cumulatively create a distinct systemic metabolic pathophysiological syndrome that predisposes patients to several diseases including breast cancer. Moreover, diabetic and obese women with breast cancer show a significant increase in mortality compared to non-obese and/or non-diabetic women. We hypothesized that these metabolic conditions incite an aggressive tumor phenotype by way of impacting tumor cell-autonomous and tumor cell non-autonomous events. In this study, we established a type 2 diabetic mouse model of triple-negative mammary carcinoma and investigated the effect of a glucose lowering therapy, metformin, on the overall tumor characteristics and immune/metabolic microenvironment. Diabetic mice exhibited larger mammary tumors that had increased adiposity with high levels of O-GlcNAc protein post-translational modification. These tumors also presented with a distinct stromal profile characterized by altered collagen architecture, increased infiltration by tumor-permissive M2 macrophages, and early metastatic seeding compared to non-diabetic/lean mice. Metformin treatment of the diabetic/obese mice effectively normalized glucose levels, reconfigured the mammary tumor milieu, and decreased metastatic seeding. Our results highlight the impact of two metabolic complications of obesity and diabetes on tumor cell attributes and showcase metformin's ability to revert tumor cell and stromal changes induced by an obese and diabetic host environment.
Collapse
Affiliation(s)
| | - Brandon J Metge
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chae-Myeong Ha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dominique C Hinshaw
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mateus S V Mota
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah C Kammerud
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tshering Lama-Sherpa
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Noha Sharafeldin
- Division of Hematology & Oncology, Dept of Medicine, UAB School of Medicine, UAB, USA; Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
12
|
Brahma MK, Ha C, Pepin ME, Mia S, Sun Z, Chatham JC, Habegger KM, Abel ED, Paterson AJ, Young ME, Wende AR. Increased Glucose Availability Attenuates Myocardial Ketone Body Utilization. J Am Heart Assoc 2020; 9:e013039. [PMID: 32750298 PMCID: PMC7792234 DOI: 10.1161/jaha.119.013039] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/05/2020] [Indexed: 02/06/2023]
Abstract
Background Perturbations in myocardial substrate utilization have been proposed to contribute to the pathogenesis of cardiac dysfunction in diabetic subjects. The failing heart in nondiabetics tends to decrease reliance on fatty acid and glucose oxidation, and increases reliance on ketone body oxidation. In contrast, little is known regarding the mechanisms mediating this shift among all 3 substrates in diabetes mellitus. Therefore, we tested the hypothesis that changes in myocardial glucose utilization directly influence ketone body catabolism. Methods and Results We examined ventricular-cardiac tissue from the following murine models: (1) streptozotocin-induced type 1 diabetes mellitus; (2) high-fat-diet-induced glucose intolerance; and transgenic inducible cardiac-restricted expression of (3) glucose transporter 4 (transgenic inducible cardiac restricted expression of glucose transporter 4); or (4) dominant negative O-GlcNAcase. Elevated blood glucose (type 1 diabetes mellitus and high-fat diet mice) was associated with reduced cardiac expression of β-hydroxybutyrate-dehydrogenase and succinyl-CoA:3-oxoacid CoA transferase. Increased myocardial β-hydroxybutyrate levels were also observed in type 1 diabetes mellitus mice, suggesting a mismatch between ketone body availability and utilization. Increased cellular glucose delivery in transgenic inducible cardiac restricted expression of glucose transporter 4 mice attenuated cardiac expression of both Bdh1 and Oxct1 and reduced rates of myocardial BDH1 activity and β-hydroxybutyrate oxidation. Moreover, elevated cardiac protein O-GlcNAcylation (a glucose-derived posttranslational modification) by dominant negative O-GlcNAcase suppressed β-hydroxybutyrate dehydrogenase expression. Consistent with the mouse models, transcriptomic analysis confirmed suppression of BDH1 and OXCT1 in patients with type 2 diabetes mellitus and heart failure compared with nondiabetic patients. Conclusions Our results provide evidence that increased glucose leads to suppression of cardiac ketolytic capacity through multiple mechanisms and identifies a potential crosstalk between glucose and ketone body metabolism in the diabetic myocardium.
Collapse
Affiliation(s)
- Manoja K. Brahma
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - Chae‐Myeong Ha
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - Mark E. Pepin
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
- Biomedical EngineeringUniversity of Alabama at BirminghamALUSA
| | - Sobuj Mia
- Medicine, Division of Cardiovascular DiseasesUniversity of Alabama at BirminghamALUSA
| | - Zhihuan Sun
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - John C. Chatham
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - Kirk M. Habegger
- Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamALUSA
| | - Evan Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIAUSA
| | - Andrew J. Paterson
- Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamALUSA
| | - Martin E. Young
- Medicine, Division of Cardiovascular DiseasesUniversity of Alabama at BirminghamALUSA
| | - Adam R. Wende
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
- Biomedical EngineeringUniversity of Alabama at BirminghamALUSA
| |
Collapse
|
13
|
Avtanski D, Pavlov VA, Tracey KJ, Poretsky L. Characterization of inflammation and insulin resistance in high-fat diet-induced male C57BL/6J mouse model of obesity. Animal Model Exp Med 2019; 2:252-258. [PMID: 31942557 PMCID: PMC6930989 DOI: 10.1002/ame2.12084] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/08/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Animal models of diet-induced obesity (DIO) are commonly used in medical research for mimicking human diseases. There is no universal animal model, and careful evaluation of variety of factors needs to be considered when designing new experiments. Here, we investigated the effect of 9 weeks high-fat diet (HFD) intervention, providing 60% energy from fat, on parameters of inflammation and insulin resistance in male C57BL/6J mice. METHODS Six weeks old mice were initiated on regular diet (RD) or HFD providing 60 kcal energy from fat for 9 weeks. Fasting blood glucose levels were measured by glucometer, and fasting plasma levels of insulin and proinflammatory cytokines by Luminex assay. Insulin sensitivity was evaluated by using QUICKI and HOMA2 indexes. RESULTS HFD mice showed ~ 40% higher body weight and ~ 20% larger abdominal circumference, due to an increase in the white adipose tissue mass. Liver examination revealed increased size and higher hepatic lipid accumulation in livers from HFD mice compared to their RD counterparts. Animals from the HFD group were characterized with significantly higher presence of crown-like structures (CLS) in WAT and higher plasma levels of proinflammatory cytokines (TNF-α, IL-6, leptin, MCP-1, PAI-1, and resistin). HFD-fed mice also demonstrated impaired insulin sensitivity (lower QUICKI, higher HOMA-insulin resistance (HOMA-IR), and lower HOMA-percent sensitivity (HOMA-%S)) index values. CONCLUSION Male C57BL/6J mice on 9 weeks HFD providing 60 kcal energy from fat display impaired insulin sensitivity and chronic inflammation, thus making this DIO mouse model appropriate for studies of early stages of obesity-related pathology.
Collapse
Affiliation(s)
- Dimiter Avtanski
- Lenox Hill HospitalFriedman Diabetes InstituteNorthwell HealthNew YorkNYUSA
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNYUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNYUSA
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNYUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNYUSA
| | - Kevin J. Tracey
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNYUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNYUSA
| | - Leonid Poretsky
- Lenox Hill HospitalFriedman Diabetes InstituteNorthwell HealthNew YorkNYUSA
- The Feinstein Institutes for Medical ResearchNorthwell HealthManhassetNYUSA
- Donald and Barbara Zucker School of Medicine at Hofstra/NorthwellHempsteadNYUSA
| |
Collapse
|
14
|
Peng S, Wang Y, Zhou Y, Ma T, Wang Y, Li J, Huang F, Kou J, Qi L, Liu B, Liu K. Rare ginsenosides ameliorate lipid overload-induced myocardial insulin resistance via modulating metabolic flexibility. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 58:152745. [PMID: 31005715 DOI: 10.1016/j.phymed.2018.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Rare ginsenosides are found in ginseng and notoginseng, two medicinal plants widely used in China for treatment of cardiovascular diseases and type 2 diabetes. However, their pharmacological studies regarding myocardial fuel metabolism and insulin signaling are not clear. PURPOSE To explore the effect of a rare ginsenoside-standardized extract (RGSE), derived from steamed notoginseng, on cardiac fuel metabolism and insulin signaling. STUDY DESIGN We used palmitic acid (PA) to treat H9c2 cells in vitro and high fat diet (HFD) to mice to induce insulin resistance in vivo. METHODS In vitro, differentiated H9c2 cells were pretreated with RGSE, metformin, mildronate or dichloroacetate (DCA) and stimulated with PA. In vivo, mice were fed with HFD and received RGSE, metformin or DCA for 6 weeks. Protein expression was determined by Western blotting. Mitochondrial membrane potential (Δψm), glucose uptake and reactive oxygen species (ROS) production were measured by fluorescence labeling. Other assessments including oxygen consumption rate (OCR) were also performed. RESULTS RGSE prevented PA-induced decrease in pyruvate dehydrogenase (PDH) activity and increase in carnitine palmitoyltransferase 1 (CPT1) expression, and ameliorated insulin-mediated glucose uptake and utilization in H9c2 cells. Metformin and mildronate exhibited similar effects. In vivo, RGSE counteracted HFD-induced increase in myocardial expression of p-PDH and CPT1 and ameliorated cardiac insulin signaling. Metformin and DCA also showed beneficial effects. Further study showed that RGSE decreased OCR and mitochondrial complex I activity in PA-treated H9c2 cells, reduced ROS production and relieved mitochondrial oxidative stress, thus decreased serine phosphorylation in IRS-1. CONCLUSION RGSE ameliorated myocardial insulin sensitivity under conditions of lipid overload, which was tightly associated with the decrease in mitochondrial oxidative stress via modulating glucose and fatty acid oxidation.
Collapse
Affiliation(s)
- Shuang Peng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Yilei Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Ying Zhou
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Tingting Ma
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Yapu Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Jia Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjiaxiang, Nanjing 210009, China
| | - Fang Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Lianwen Qi
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjiaxiang, Nanjing 210009, China
| | - Baolin Liu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China
| | - Kang Liu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Longmian Road, Nanjing 211198, China.
| |
Collapse
|
15
|
Pedersen TM, Boardman NT, Hafstad AD, Aasum E. Isolated perfused working hearts provide valuable additional information during phenotypic assessment of the diabetic mouse heart. PLoS One 2018; 13:e0204843. [PMID: 30273374 PMCID: PMC6166959 DOI: 10.1371/journal.pone.0204843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022] Open
Abstract
Although murine models for studying the development of cardiac dysfunction in diabetes mellitus are well established, their reported cardiac phenotypes vary. These reported divergences may, in addition to the severity of different models, also be linked to the methods used for cardiac functional assessment. In the present study, we examined the functional changes using conventional transthoracic echocardiography (in vivo) and isolated heart perfusion techniques (ex vivo), in hearts from two mouse models; one with an overt type 2 diabetes (the db/db mouse) and one with a prediabetic state, where obesity was induced by a high-fat diet (HFD). Analysis of left ventricular function in the isolated working hearts from HFD-fed mice, suggested that these hearts develop diastolic dysfunction with preserved systolic function. Accordingly, in vivo examination demonstrated maintained systolic function, but we did not find parameters of diastolic function to be altered. In db/db mice, ex vivo working hearts showed both diastolic and systolic dysfunction. Although in vivo functional assessment revealed signs of diastolic dysfunction, the hearts did not display reduced systolic function. The contrasting results between ex vivo and in vivo function could be due to systemic changes that may sustain in vivo function, or a lack of sensitivity using conventional transthoracic echocardiography. Thus, this study demonstrates that the isolated perfused working heart preparation provides unique additional information related to the development of cardiomyopathy, which might otherwise go unnoticed when only using conventional echocardiographic assessment.
Collapse
Affiliation(s)
- Tina M. Pedersen
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Neoma T. Boardman
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Anne D. Hafstad
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ellen Aasum
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
- * E-mail:
| |
Collapse
|
16
|
Knockout of CNR1 prevents metabolic stress-induced cardiac injury through improving insulin resistance (IR) injury and endoplasmic reticulum (ER) stress by promoting AMPK-alpha activation. Biochem Biophys Res Commun 2018; 503:744-751. [DOI: 10.1016/j.bbrc.2018.06.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 01/10/2023]
|
17
|
LNK deficiency aggravates palmitate-induced preadipocyte apoptosis. Biochem Biophys Res Commun 2017; 490:91-97. [DOI: 10.1016/j.bbrc.2017.05.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 01/06/2023]
|
18
|
Han F, Zhou D, Yin X, Sun Z, Han J, Ye L, Zhao W, Zhang Y, Wang Z, Zheng L. Paeoniflorin protects diabetic mice against myocardial ischemic injury via the transient receptor potential vanilloid 1/calcitonin gene-related peptide pathway. Cell Biosci 2016; 6:37. [PMID: 27252827 PMCID: PMC4888521 DOI: 10.1186/s13578-016-0085-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/21/2016] [Indexed: 12/13/2022] Open
Abstract
Background Diabetes mellitus has multiple complications including neuropathy and increases cardiovascular events. Paeoniflorin (PF), a monoterpene glycoside, plays an essential role in neuroprotection and ischemic heart disease. In this study, we aimed to investigate the hypothesis that PF protects mice with diabetes mellitus against myocardial ischemic injury, and determine its associated mechanisms. Results Myocardial infarction (MI) was generated in the streptozotocin-mediated diabetic mice, which were pretreated with either vehicle or PF, respectively. Myocardial infarct size, myocardial enzyme, cardiac function, circulating calcitonin gene-related peptide (CGRP) concentration, histological analysis and the expression of associated molecules were determined and compared among different experimental groups. Compared to diabetic hearts pretreated with vehicle, hearts pretreated with PF exhibited less tissue damage and better CGRP concentration in serum when subjected to myocardial ischemia. Transient receptor potential vanilloid 1(TRPV1) gene knockout attenuated PF-mediated cardioprotection. Moreover, a specific Ca2+/calmodulin-dependent protein kinase (CaMK) inhibitor, KN-93, increased tissue damage and decreased CGRP activity in serum. Meanwhile, pretreated with PF increased the phosphorylation of cAMP response element binding protein (CREB). Conclusions Taken together, these findings demonstrate that PF protects diabetic mice against MI at least partially via the TRPV1/CaMK/CREB/CGRP signaling pathway.
Collapse
Affiliation(s)
- Fei Han
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Dongchen Zhou
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Xiang Yin
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Zewei Sun
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Jie Han
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Lifang Ye
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Wengting Zhao
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Yuanyuan Zhang
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Zhen Wang
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Liangrong Zheng
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| |
Collapse
|
19
|
Enhanced cartilage repair in 'healer' mice-New leads in the search for better clinical options for cartilage repair. Semin Cell Dev Biol 2016; 62:78-85. [PMID: 27130635 DOI: 10.1016/j.semcdb.2016.04.018] [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: 03/08/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Adult articular cartilage has a poor capacity to undergo intrinsic repair. Current strategies for the repair of large cartilage defects are generally unsatisfactory because the restored cartilage does not have the same resistance to biomechanical loading as authentic articular cartilage and degrades over time. Recently, an exciting new research direction, focused on intrinsic cartilage regeneration rather than fibrous repair by external means, has emerged. This review explores the new findings in this rapidly moving field as they relate to the clinical goal of restoration of structurally robust, stable and non-fibrous articular cartilage following injury.
Collapse
|
20
|
Heydemann A. An Overview of Murine High Fat Diet as a Model for Type 2 Diabetes Mellitus. J Diabetes Res 2016; 2016:2902351. [PMID: 27547764 PMCID: PMC4983380 DOI: 10.1155/2016/2902351] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a worldwide epidemic, which by all predictions will only increase. To help in combating the devastating array of phenotypes associated with T2DM a highly reproducible and human disease-similar mouse model is required for researchers. The current options are genetic manipulations to cause T2DM symptoms or diet induced obesity and T2DM symptoms. These methods to model human T2DM have their benefits and their detractions. As far as modeling the majority of T2DM cases, HFD establishes the proper etiological, pathological, and treatment options. A limitation of HFD is that it requires months of feeding to achieve the full spectrum of T2DM symptoms and no standard protocol has been established. This paper will attempt to rectify the last limitation and argue for a standard group of HFD protocols and standard analysis procedures.
Collapse
Affiliation(s)
- Ahlke Heydemann
- The University of Illinois at Chicago, Chicago, IL 60612, USA
- The Center for Cardiovascular Research, Chicago, IL 60612, USA
- *Ahlke Heydemann:
| |
Collapse
|