1
|
Berezin OO, Berezina TA, Hoppe UC, Lichtenauer M, Berezin AE. Diagnostic and predictive abilities of myokines in patients with heart failure. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:45-98. [PMID: 39059994 DOI: 10.1016/bs.apcsb.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Myokines are defined as a heterogenic group of numerous cytokines, peptides and metabolic derivates, which are expressed, synthesized, produced, and released by skeletal myocytes and myocardial cells and exert either auto- and paracrine, or endocrine effects. Previous studies revealed that myokines play a pivotal role in mutual communications between skeletal muscles, myocardium and remote organs, such as brain, vasculature, bone, liver, pancreas, white adipose tissue, gut, and skin. Despite several myokines exert complete divorced biological effects mainly in regulation of skeletal muscle hypertrophy, residential cells differentiation, neovascularization/angiogenesis, vascular integrity, endothelial function, inflammation and apoptosis/necrosis, attenuating ischemia/hypoxia and tissue protection, tumor growth and malignance, for other occasions, their predominant effects affect energy homeostasis, glucose and lipid metabolism, adiposity, muscle training adaptation and food behavior. Last decade had been identified 250 more myokines, which have been investigating for many years further as either biomarkers or targets for heart failure management. However, only few myokines have been allocated to a promising tool for monitoring adverse cardiac remodeling, ischemia/hypoxia-related target-organ dysfunction, microvascular inflammation, sarcopenia/myopathy and prediction for poor clinical outcomes among patients with HF. This we concentrate on some most plausible myokines, such as myostatin, myonectin, brain-derived neurotrophic factor, muslin, fibroblast growth factor 21, irisin, leukemia inhibitory factor, developmental endothelial locus-1, interleukin-6, nerve growth factor and insulin-like growth factor-1, which are suggested to be useful biomarkers for HF development and progression.
Collapse
Affiliation(s)
- Oleksandr O Berezin
- Luzerner Psychiatrie AG, Department of Senior Psychiatrie, St. Urban, Switzerland
| | - Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, Ukraine
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
2
|
Ito M, Shibata R, Ohashi K, Otaka N, Yamaguchi S, Ogawa H, Enomoto T, Masutomi T, Murohara T, Ouchi N. Omentin Modulates Chronic Cardiac Remodeling After Myocardial Infarction. Circ Rep 2023; 5:46-54. [PMID: 36818520 PMCID: PMC9908527 DOI: 10.1253/circrep.cr-22-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/25/2022] [Accepted: 12/26/2022] [Indexed: 01/19/2023] Open
Abstract
Background: Omentin, a circulating adipokine, is downregulated in complications of obesity, including heart disease. Here, we investigated whether omentin modulates adverse cardiac remodeling in mice after myocardial infarction (MI). Methods and Results: Transgenic mice expressing the human omentin gene in fat tissue (OMT-Tg) and wild-type (WT) mice were subjected to permanent ligation of the left anterior descending coronary artery (LAD) to induce MI. OMT-Tg mice had a higher survival rate after permanent LAD ligation than WT mice. Moreover, OMT-Tg mice had lower heart weight/body weight (HW/BW) and lung weight/body weight (LW/BW) ratios at 4 weeks after coronary artery ligation compared with WT mice. OMT-Tg mice also showed decreased left ventricular diastolic diameter (LVDd) and increased fractional shortening (%FS) following MI. Moreover, an increase in capillary density in the infarct border zone and a decrease in myocardial apoptosis, myocyte hypertrophy, and interstitial fibrosis in the remote zone following MI, were more prevalent in OMT-Tg than WT mice. Finally, intravenous administration of adenoviral vectors expressing human omentin to WT mice after MI resulted in decreases in HW/BW, LW/BW, and LVDd, and an increase in %FS. Conclusions: Our findings document that human omentin prevents pathological cardiac remodeling after chronic ischemia, suggesting that omentin represents a potential therapeutic molecule for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Masanori Ito
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Shukuro Yamaguchi
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Hayato Ogawa
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Takashi Enomoto
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Tomohiro Masutomi
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
3
|
Li M, Li J, Song Y. Hsa_Circ_0134426 Attenuates the Malignant Biological Behaviors of Multiple Myeloma by Suppressing miR-146b-3p to Upregulate NDNF. Mol Biotechnol 2022:10.1007/s12033-022-00618-6. [DOI: 10.1007/s12033-022-00618-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022]
|
4
|
Li X, Zhu X, Li B, Xia B, Tang H, Hu J, Ying R. Loss of α7nAChR enhances endothelial-to-mesenchymal transition after myocardial infarction via NF-κB activation. Exp Cell Res 2022; 419:113300. [PMID: 35926661 DOI: 10.1016/j.yexcr.2022.113300] [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: 01/06/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022]
Abstract
The myocardial fibrosis in response to myocardial infarction (MI) is closely related to the dysbalance of endothelial-to-mesenchymal transition (EndMT). Although numerous reports indicate that α7 nicotinic acetylcholine receptor (α7nAChR) activates the cholinergic anti-inflammatory pathway (CAP) to regulate the magnitude of inflammatory responses, the role of α7nAChR in myocardial fibrosis, as well as the underlying mechanisms, have not been elucidated. In this study, we evaluated cardiac function, fibrosis, and EndMT signaling using a mouse model of MI and interleukin (IL)-1β-induced human cardiac microvascular endothelial cells (HCMECs). In vivo, α7nAChR deletion increased cardiac dysfunction, exacerbated the cardiac inflammatory response, and NF-κB activation, and enhanced EndMT, as shown by higher expression levels of fibroblast markers (FSP-1, α-SMA, collagen I, Snail) and decreased levels of the FGFR1, glucocorticoid receptor (GR) and endothelial marker (CD31) compared to wild-type mice. In vitro, the pharmacological activation of α7nAChR with PNU282987 significantly inhibited IL-1β-induced EndMT, as shown by a reduced transition to the fibroblast-like phenotype and the expression of fibrotic markers. Moreover, the IL-1β-mediated activation of NF-κB pathway was suppressed by PNU282987. This anti-EndMT effect of α7nAChR was associated with regulation of Snail. Furthermore, Western blot analysis further revealed that the GR antagonist RU38486 could partially counteract the effect of PNU282987 on NF-κB expression. In conclusion, our results show that α7nAChR is involved in cardiac fibrosis by inhibiting EndMT, providing a novel approach to the treatment of MI.
Collapse
Affiliation(s)
- Xuelian Li
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, 266011, China.
| | - Xianjie Zhu
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, Shandong, 266011, China.
| | - Bingong Li
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, 266011, China; Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Baohua Xia
- Department of Clinical Skills Training Center, Qingdao Municipal Hospital, Qingdao, Shandong, 266011, China.
| | - Huaiguang Tang
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, 266011, China.
| | - Jinxing Hu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Ru Ying
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
5
|
Akiyama T, Seidel CW, Gibson MC. The feedback regulator nord controls Dpp/BMP signaling via extracellular interaction with dally in the Drosophila wing. Dev Biol 2022; 488:91-103. [DOI: 10.1016/j.ydbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/27/2022]
|
6
|
Ozaki Y, Ohashi K, Otaka N, Ogawa H, Kawanishi H, Takikawa T, Fang L, Tatsumi M, Takefuji M, Enomoto T, Darwish M, Iijima Y, Iijima T, Murohara T, Ouchi N. Neuron-derived neurotrophic factor protects against dexamethasone-induced skeletal muscle atrophy. Biochem Biophys Res Commun 2022; 593:5-12. [DOI: 10.1016/j.bbrc.2022.01.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/09/2022] [Indexed: 12/20/2022]
|
7
|
Yang S, Wu X, Daoutidou EI, Zhang Y, Shimell M, Chuang KH, Peterson AJ, O'Connor MB, Zheng X. The NDNF-like factor Nord is a Hedgehog-induced extracellular BMP modulator that regulates Drosophila wing patterning and growth. eLife 2022; 11:e73357. [PMID: 35037619 PMCID: PMC8856659 DOI: 10.7554/elife.73357] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/15/2022] [Indexed: 11/23/2022] Open
Abstract
Hedgehog (Hh) and Bone Morphogenetic Proteins (BMPs) pattern the developing Drosophila wing by functioning as short- and long-range morphogens, respectively. Here, we show that a previously unknown Hh-dependent mechanism fine-tunes the activity of BMPs. Through genome-wide expression profiling of the Drosophila wing imaginal discs, we identify nord as a novel target gene of the Hh signaling pathway. Nord is related to the vertebrate Neuron-Derived Neurotrophic Factor (NDNF) involved in congenital hypogonadotropic hypogonadism and several types of cancer. Loss- and gain-of-function analyses implicate Nord in the regulation of wing growth and proper crossvein patterning. At the molecular level, we present biochemical evidence that Nord is a secreted BMP-binding protein and localizes to the extracellular matrix. Nord binds to Decapentaplegic (Dpp) or the heterodimer Dpp-Glass-bottom boat (Gbb) to modulate their release and activity. Furthermore, we demonstrate that Nord is a dosage-dependent BMP modulator, where low levels of Nord promote and high levels inhibit BMP signaling. Taken together, we propose that Hh-induced Nord expression fine-tunes both the range and strength of BMP signaling in the developing Drosophila wing.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Euphrosyne I Daoutidou
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Ya Zhang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - MaryJane Shimell
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Kun-Han Chuang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Aidan J Peterson
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Michael B O'Connor
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| |
Collapse
|
8
|
Vladimirsky VE, Vladimirsky EV, Lunina AN, Fesyun AD, Rachin AP, Lebedeva OD, Yakovlev MY, Tubekova MA. [Molecular mechanisms of adaptive and therapeutic effects of physical activity in patients with cardiovascular diseases]. VOPROSY KURORTOLOGII, FIZIOTERAPII, I LECHEBNOI FIZICHESKOI KULTURY 2022; 99:69-77. [PMID: 35485663 DOI: 10.17116/kurort20229902169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Physical activity is one of the main components of the rehabilitation of patients with cardiovascular disease (CVD). As shown by practice and the results of evidence-based studies, the beneficial effects of physical activity on disease outcomes in a number of cardiac nosologies are comparable to drug treatment. This gives the doctor another tool to influence the unfavorable epidemiological situation in developed countries with the spread of diseases of the cardiovascular system and CVD mortality. Reliable positive results of cardiorehabilitation (CR) were obtained using various methods. The goal of CR is to restore the optimal physiological, psychological and professional status, reduce the risk of CVD and mortality. In most current CVD guidelines worldwide, cardiac rehabilitation is a Class I recommendation. The molecular mechanisms described in the review, initiated by physical activity, underlie the multifactorial effect of the latter on the function of the cardiovascular system and the course of cardiac diseases. Physical exercise is an important component of the therapeutic management of patients with CVD, which is supported by the results of a meta-analysis of 63 studies associated with various forms of aerobic exercise of varying intensity (from 50 to 95% VO2) for 1 to 47 months, which showed that CR based on physical exercise improves cardiorespiratory endurance. Knowledge of the molecular basis of the influence of physical activity makes it possible to use biochemical markers to assess the effectiveness of rehabilitation programs.
Collapse
Affiliation(s)
| | | | - A N Lunina
- Wagner Perm State Medical University, Perm, Russia
| | - A D Fesyun
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - A P Rachin
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - O D Lebedeva
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - M Yu Yakovlev
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - M A Tubekova
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| |
Collapse
|
9
|
Borrelli MA, Turnquist HR, Little SR. Biologics and their delivery systems: Trends in myocardial infarction. Adv Drug Deliv Rev 2021; 173:181-215. [PMID: 33775706 PMCID: PMC8178247 DOI: 10.1016/j.addr.2021.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/14/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is the leading cause of death around the world, in which myocardial infarction (MI) is a precipitating event. However, current therapies do not adequately address the multiple dysregulated systems following MI. Consequently, recent studies have developed novel biologic delivery systems to more effectively address these maladies. This review utilizes a scientometric summary of the recent literature to identify trends among biologic delivery systems designed to treat MI. Emphasis is placed on sustained or targeted release of biologics (e.g. growth factors, nucleic acids, stem cells, chemokines) from common delivery systems (e.g. microparticles, nanocarriers, injectable hydrogels, implantable patches). We also evaluate biologic delivery system trends in the entire regenerative medicine field to identify emerging approaches that may translate to the treatment of MI. Future developments include immune system targeting through soluble factor or chemokine delivery, and the development of advanced delivery systems that facilitate the synergistic delivery of biologics.
Collapse
Affiliation(s)
- Matthew A Borrelli
- Department of Chemical Engineering, University of Pittsburgh, 940 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, USA.
| | - Heth R Turnquist
- Starzl Transplantation Institute, 200 Darragh St, Pittsburgh, PA 15213, USA; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| | - Steven R Little
- Department of Chemical Engineering, University of Pittsburgh, 940 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Clinical and Translational Science, University of Pittsburgh, Forbes Tower, Suite 7057, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, USA; Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Pharmaceutical Science, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15213, USA; Department of Ophthalmology, University of Pittsburgh, 203 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
10
|
Cavus O, Williams J, Musa H, El Refaey M, Gratz D, Shaheen R, Schwieterman NA, Koenig S, Antwi-Boasiako S, Young LJ, Xu X, Han M, Wold LE, Hund TJ, Mohler PJ, Bradley EA. Giant ankyrin-G regulates cardiac function. J Biol Chem 2021; 296:100507. [PMID: 33675749 PMCID: PMC8040283 DOI: 10.1016/j.jbc.2021.100507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/30/2022] Open
Abstract
Cardiovascular disease (CVD) remains the most common cause of adult morbidity and mortality in developed nations. As a result, predisposition for CVD is increasingly important to understand. Ankyrins are intracellular proteins required for the maintenance of membrane domains. Canonical ankyrin-G (AnkG) has been shown to be vital for normal cardiac function, specifically cardiac excitability, via targeting and regulation of the cardiac voltage-gated sodium channel. Noncanonical (giant) AnkG isoforms play a key role in neuronal membrane biogenesis and excitability, with evidence for human neurologic disease when aberrant. However, the role of giant AnkG in cardiovascular tissue has yet to be explored. Here, we identify giant AnkG in the myocardium and identify that it is enriched in 1-week-old mice. Using a new mouse model lacking giant AnkG expression in myocytes, we identify that young mice displayed a dilated cardiomyopathy phenotype with aberrant electrical conduction and enhanced arrhythmogenicity. Structural and electrical dysfunction occurred at 1 week of age, when giant AnkG was highly expressed and did not appreciably change in adulthood until advanced age. At a cellular level, loss of giant AnkG results in delayed and early afterdepolarizations. However, surprisingly, giant AnkG cKO myocytes display normal INa, but abnormal myocyte contractility, suggesting unique roles of the large isoform in the heart. Finally, transcript analysis provided evidence for unique pathways that may contribute to the structural and electrical findings shown in giant AnkG cKO animals. In summary, we identify a critical role for giant AnkG that adds to the diversity of ankyrin function in the heart.
Collapse
Affiliation(s)
- Omer Cavus
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Jordan Williams
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Hassan Musa
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Mona El Refaey
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Dan Gratz
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Rebecca Shaheen
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Neill A Schwieterman
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Sara Koenig
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Steve Antwi-Boasiako
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Lindsay J Young
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Xianyao Xu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Mei Han
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Loren E Wold
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Thomas J Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; Department of Internal Medicine/Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Mohler
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Internal Medicine/Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Elisa A Bradley
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA; Department of Internal Medicine/Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
11
|
Takikawa T, Ohashi K, Ogawa H, Otaka N, Kawanishi H, Fang L, Ozaki Y, Eguchi S, Tatsumi M, Takefuji M, Murohara T, Ouchi N. Adipolin/C1q/Tnf-related protein 12 prevents adverse cardiac remodeling after myocardial infarction. PLoS One 2020; 15:e0243483. [PMID: 33275602 PMCID: PMC7717554 DOI: 10.1371/journal.pone.0243483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/21/2020] [Indexed: 11/19/2022] Open
Abstract
Background Myocardial infarction (MI) is a leading cause of death worldwide. We previously identified adipolin, also known as C1q/Tnf-related protein 12, as an anti-inflammatory adipokine with protective features against metabolic and vascular disorders. Here, we investigated the effect of adipolin on myocardial remodeling in a mouse model of MI. Methods Male adipolin-knockout (APL-KO) and wild-type (WT) mice were subjected to the permanent ligation of the left anterior descending coronary artery to create MI. Results APL-KO mice exhibited increased ratios of heart weight/body weight and lung weight/body weight after MI compared with WT mice. APL-KO mice showed increased left ventricular diastolic diameter and decreased fractional shortening after MI compared with WT mice. APL-KO mice exhibited increased expression of pro-inflammatory mediators and enhanced cardiomyocyte apoptosis in the post-MI hearts compared with WT mice. Systemic administration of adenoviral vectors expressing adipolin to WT mice after MI surgery improved left ventricular contractile dysfunction and reduced cardiac expression of pro-inflammatory genes. Treatment of cultured cardiomyocytes with adipolin protein reduced lipopolysaccharide-induced expression of pro-inflammatory mediators and hypoxia-induced apoptosis. Treatment with adipolin protein increased Akt phosphorylation in cardiomyocytes. Inhibition of PI3 kinase/Akt signaling reversed the anti-inflammatory and anti-apoptotic effects of adipolin in cardiomyocytes. Conclusion Our data indicate that adipolin ameliorates pathological remodeling of myocardium after MI, at least in part, by its ability to reduce myocardial inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Tomonobu Takikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail: (KO); (NO)
| | - Hayato Ogawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kawanishi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Lixin Fang
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuta Ozaki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shunsuke Eguchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Minako Tatsumi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail: (KO); (NO)
| |
Collapse
|
12
|
Messina A, Pulli K, Santini S, Acierno J, Känsäkoski J, Cassatella D, Xu C, Casoni F, Malone SA, Ternier G, Conte D, Sidis Y, Tommiska J, Vaaralahti K, Dwyer A, Gothilf Y, Merlo GR, Santoni F, Niederländer NJ, Giacobini P, Raivio T, Pitteloud N. Neuron-Derived Neurotrophic Factor Is Mutated in Congenital Hypogonadotropic Hypogonadism. Am J Hum Genet 2020; 106:58-70. [PMID: 31883645 DOI: 10.1016/j.ajhg.2019.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022] Open
Abstract
Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by infertility and the absence of puberty. Defects in GnRH neuron migration or altered GnRH secretion and/or action lead to a severe gonadotropin-releasing hormone (GnRH) deficiency. Given the close developmental association of GnRH neurons with the olfactory primary axons, CHH is often associated with anosmia or hyposmia, in which case it is defined as Kallmann syndrome (KS). The genetics of CHH are heterogeneous, and >40 genes are involved either alone or in combination. Several CHH-related genes controlling GnRH ontogeny encode proteins containing fibronectin-3 (FN3) domains, which are important for brain and neural development. Therefore, we hypothesized that defects in other FN3-superfamily genes would underlie CHH. Next-generation sequencing was performed for 240 CHH unrelated probands and filtered for rare, protein-truncating variants (PTVs) in FN3-superfamily genes. Compared to gnomAD controls the CHH cohort was statistically enriched for PTVs in neuron-derived neurotrophic factor (NDNF) (p = 1.40 × 10-6). Three heterozygous PTVs (p.Lys62∗, p.Tyr128Thrfs∗55, and p.Trp469∗, all absent from the gnomAD database) and an additional heterozygous missense mutation (p.Thr201Ser) were found in four KS probands. Notably, NDNF is expressed along the GnRH neuron migratory route in both mouse embryos and human fetuses and enhances GnRH neuron migration. Further, knock down of the zebrafish ortholog of NDNF resulted in altered GnRH migration. Finally, mice lacking Ndnf showed delayed GnRH neuron migration and altered olfactory axonal projections to the olfactory bulb; both results are consistent with a role of NDNF in GnRH neuron development. Altogether, our results highlight NDNF as a gene involved in the GnRH neuron migration implicated in KS.
Collapse
Affiliation(s)
- Andrea Messina
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Kristiina Pulli
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Sara Santini
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - James Acierno
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland; Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Johanna Känsäkoski
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Daniele Cassatella
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland; Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Cheng Xu
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Filippo Casoni
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Unité 1172 Lille, 59045 Lille, France; Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy, Milan 20132, Italy; Università Vita-Salute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Samuel A Malone
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Unité 1172 Lille, 59045 Lille, France
| | - Gaetan Ternier
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Unité 1172 Lille, 59045 Lille, France
| | - Daniele Conte
- Department of Molecular Biotechnology and Health Science, University of Torino, 10126 Torino, Italy
| | - Yisrael Sidis
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Johanna Tommiska
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Andrew Dwyer
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Yoav Gothilf
- Department of Neurobiology, George S. Wise Faculty of Life Sciences and Sagol School of Neurosciences, University of Tel Aviv, Tel Aviv 69978, Israel
| | - Giorgio R Merlo
- Department of Molecular Biotechnology and Health Science, University of Torino, 10126 Torino, Italy
| | - Federico Santoni
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nicolas J Niederländer
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Paolo Giacobini
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Unité 1172 Lille, 59045 Lille, France
| | - Taneli Raivio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; Pediatric Research Center, New Children's Hospital, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland.
| |
Collapse
|
13
|
Zhang Y, Wu X, Kai Y, Lee CH, Cheng F, Li Y, Zhuang Y, Ghaemmaghami J, Chuang KH, Liu Z, Meng Y, Keswani M, Gough NR, Wu X, Zhu W, Tzatsos A, Peng W, Seto E, Sotomayor EM, Zheng X. Secretome profiling identifies neuron-derived neurotrophic factor as a tumor-suppressive factor in lung cancer. JCI Insight 2019; 4:129344. [PMID: 31852841 DOI: 10.1172/jci.insight.129344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
Clinical and preclinical studies show tissue-specific differences in tumorigenesis. Tissue specificity is controlled by differential gene expression. We prioritized genes that encode secreted proteins according to their preferential expression in normal lungs to identify candidates associated with lung cancer. Indeed, most of the lung-enriched genes identified in our analysis have known or suspected roles in lung cancer. We focused on the gene encoding neuron-derived neurotrophic factor (NDNF), which had not yet been associated with lung cancer. We determined that NDNF was preferentially expressed in the normal adult lung and that its expression was decreased in human lung adenocarcinoma and a mouse model of this cancer. Higher expression of NDNF was associated with better clinical outcome of patients with lung adenocarcinoma. Purified NDNF inhibited proliferation of lung cancer cells, whereas silencing NDNF promoted tumor cell growth in culture and in xenograft models. We determined that NDNF is downregulated through DNA hypermethylation near CpG island shores in human lung adenocarcinoma. Furthermore, the lung cancer-related DNA hypermethylation sites corresponded to the methylation sites that occurred in tissues with low NDNF expression. Thus, by analyzing the tissue-specific secretome, we identified a tumor-suppressive factor, NDNF, which is associated with patient outcomes in lung adenocarcinoma.
Collapse
Affiliation(s)
- Ya Zhang
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Xuefeng Wu
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yan Kai
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Department of Physics, George Washington University Columbian College of Arts and Sciences, Washington, DC, USA
| | - Chia-Han Lee
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Department of Biochemistry and Molecular Medicine
| | - Fengdong Cheng
- GW Cancer Center and.,Division of Hematology and Oncology, Department of Medicine, and
| | - Yixuan Li
- GW Cancer Center and.,Department of Biochemistry and Molecular Medicine
| | - Yongbao Zhuang
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Javid Ghaemmaghami
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Kun-Han Chuang
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Zhuo Liu
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yunxiao Meng
- GW Cancer Center and.,Department of Biochemistry and Molecular Medicine
| | - Meghana Keswani
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Nancy R Gough
- Center for Translational Medicine, Department of Surgery, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Xiaojun Wu
- Department of Pathology, Johns Hopkins Sibley Memorial Hospital, Washington, DC, USA.,Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wenge Zhu
- GW Cancer Center and.,Department of Biochemistry and Molecular Medicine
| | - Alexandros Tzatsos
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Weiqun Peng
- GW Cancer Center and.,Department of Physics, George Washington University Columbian College of Arts and Sciences, Washington, DC, USA
| | - Edward Seto
- GW Cancer Center and.,Department of Biochemistry and Molecular Medicine
| | - Eduardo M Sotomayor
- GW Cancer Center and.,Division of Hematology and Oncology, Department of Medicine, and
| | - Xiaoyan Zheng
- GW Cancer Center and.,Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
14
|
Wang Z, Wang SP, Shao Q, Li PF, Sun Y, Luo LZ, Yan XQ, Fan ZY, Hu J, Zhao J, Hang PZ, Du ZM. Brain-derived neurotrophic factor mimetic, 7,8-dihydroxyflavone, protects against myocardial ischemia by rebalancing optic atrophy 1 processing. Free Radic Biol Med 2019; 145:187-197. [PMID: 31574344 DOI: 10.1016/j.freeradbiomed.2019.09.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/09/2023]
Abstract
Brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) pathway is associated with ischemic heart diseases (IHD). 7,8-dihydroxyflavone (7,8-DHF), BDNF mimetic, is a potent agonist of TrkB. We aimed to investigate the effects and the underlying mechanisms of 7,8-DHF on cardiac ischemia. Myocardial ischemic mouse model was induced by ligation of left anterior descending coronary artery. 7,8-DHF (5 mg/kg) was administered intraperitoneally two days after ischemia for four weeks. Echocardiography, HE staining and transmission electron microscope were used to examine the function, histology and ultrastructure of the heart. H9c2 cells were treated with hydrogen peroxide (H2O2), 7,8-DHF or TrkB inhibitor ANA-12. The effects of 7,8-DHF on cell viability, mitochondrial membrane potential (MMP) and mitochondrial superoxide generation were examined. Furthermore, mitochondrial fission and protein expression of mitochondrial dynamics (Mfn2 [mitofusin 2], OPA1 [optic atrophy 1], Drp1 [dynamin-related protein 1] and Fis-1 [fission 1]) was detected by mitotracker green staining and western blot, respectively. 7,8-DHF attenuated cardiac dysfunction and cardiomyocyte abnormality of myocardial ischemic mice. Moreover, 7,8-DHF increased cell viability and reduced cell death accompanied by improving MMP, inhibiting mitochondrial superoxide and preventing excessive mitochondrial fission of H2O2-treated H9c2 cells. The cytoprotective effects of 7,8-DHF were antagonized by ANA-12. Mechanistically, 7,8-DHF repressed OMA1-dependent conversion of L-OPA1 into S-OPA1, which was abolished by Akt inhibitor. In conclusion, 7,8-DHF protects against cardiac ischemic injury by inhibiting the proteolytic cleavage of OPA1. These findings provide a novel pharmacological effect of 7,8-DHF on mitochondrial dynamics and a new potential target for IHD.
Collapse
Affiliation(s)
- Zhen Wang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Shi-Peng Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Qun Shao
- Department of Cardiology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Pei-Feng Li
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Yue Sun
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Lan-Zi Luo
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xiu-Qing Yan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Zi-Yi Fan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Juan Hu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Jing Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University (Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University), Harbin, 150001, China.
| | - Peng-Zhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
| | - Zhi-Min Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
15
|
Yang K, Song HF, He S, Yin WJ, Fan XM, Ru F, Gong H, Zhai XY, Zhang J, Peng ZX, Xi GX, Xie J, Li RK. Effect of neuron-derived neurotrophic factor on rejuvenation of human adipose-derived stem cells for cardiac repair after myocardial infarction. J Cell Mol Med 2019; 23:5981-5993. [PMID: 31287219 PMCID: PMC6714174 DOI: 10.1111/jcmm.14456] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022] Open
Abstract
The decline of cell function caused by ageing directly impacts the therapeutic effects of autologous stem cell transplantation for heart repair. The aim of this study was to investigate whether overexpression of neuron‐derived neurotrophic factor (NDNF) can rejuvenate the adipose‐derived stem cells in the elderly and such rejuvenated stem cells can be used for cardiac repair. Human adipose‐derived stem cells (hADSCs) were obtained from donors age ranged from 17 to 92 years old. The effects of age on the biological characteristics of hADSCs and the expression of ageing‐related genes were investigated. The effects of transplantation of NDNF over‐expression stem cells on heart repair after myocardial infarction (MI) in adult mice were investigated. The proliferation, migration, adipogenic and osteogenic differentiation of hADSCs inversely correlated with age. The mRNA and protein levels of NDNF were significantly decreased in old (>60 years old) compared to young hADSCs (<40 years old). Overexpression of NDNF in old hADSCs significantly improved their proliferation and migration capacity in vitro. Transplantation of NDNF‐overexpressing old hADSCs preserved cardiac function through promoting angiogenesis on MI mice. NDNF rejuvenated the cellular function of aged hADSCs. Implantation of NDNF‐rejuvenated hADSCs improved angiogenesis and cardiac function in infarcted mouse hearts.
Collapse
Affiliation(s)
- Kun Yang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China.,Department of Endocrinology, Shanxi Dayi Hospital affiliated to Shanxi Medical University, Taiyuan, China
| | - Hui-Fang Song
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China.,Department of Anatomy, Shanxi Medical University, Taiyuan, China
| | - Sheng He
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China.,Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Wen-Juan Yin
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Xue-Mei Fan
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China.,Department of Endocrinology, Shanxi Dayi Hospital affiliated to Shanxi Medical University, Taiyuan, China
| | - Feng Ru
- Department of Urology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hui Gong
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Xiao-Yan Zhai
- Department of Anatomy, Shanxi University of Chinese Medicine, Yuci, China
| | - Jie Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Ze-Xu Peng
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Guang-Xia Xi
- Department of Endocrinology, Shanxi Dayi Hospital affiliated to Shanxi Medical University, Taiyuan, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Pinckard K, Baskin KK, Stanford KI. Effects of Exercise to Improve Cardiovascular Health. Front Cardiovasc Med 2019; 6:69. [PMID: 31214598 PMCID: PMC6557987 DOI: 10.3389/fcvm.2019.00069] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022] Open
Abstract
Obesity is a complex disease that affects whole body metabolism and is associated with an increased risk of cardiovascular disease (CVD) and Type 2 diabetes (T2D). Physical exercise results in numerous health benefits and is an important tool to combat obesity and its co-morbidities, including cardiovascular disease. Exercise prevents both the onset and development of cardiovascular disease and is an important therapeutic tool to improve outcomes for patients with cardiovascular disease. Some benefits of exercise include enhanced mitochondrial function, restoration and improvement of vasculature, and the release of myokines from skeletal muscle that preserve or augment cardiovascular function. In this review we will discuss the mechanisms through which exercise promotes cardiovascular health.
Collapse
Affiliation(s)
| | | | - Kristin I. Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
17
|
Xia L, Li S, Liu Y, Huang Y, Ni B, Wan L, Mei H, Li X, Cai Z, Li Z. NDNF inhibits the migration and invasion of human renal cancer cells through epithelial-mesenchymal transition. Oncol Lett 2019; 17:2969-2975. [PMID: 30867731 DOI: 10.3892/ol.2019.9937] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 11/21/2018] [Indexed: 11/06/2022] Open
Abstract
Neuron-derived neurotrophic factor (NDNF) is a glycosylated, disulfide-bonded secretory protein that contains a fibronectin type III domain. NDNF has been identified as a neurotrophic factor; however, its role in carcinogenesis has not yet been identified. To investigate the expression and role of NDNF in carcinogenesis, the expression of NDNF in human Renal cell carcinoma (RCC) cell lines and tissues was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. Cell proliferation was investigated using CCK-8 and colony formation assays, and the cell invasion and immigration capacity was evaluated using the transwell assay. The results demonstrated that NDNF expression was downregulated in RCC cell lines and RCC tissues. Restoring NDNF expression significantly inhibited the proliferation, migration and invasion of RCC cells. The study also demonstrated that the inhibitory effect of NDNF on invasive ability was mediated by suppressing the epithelial-mesenchymal transition (EMT) in RCC cells. NDNF may therefore be considered an important regulator of EMT in RCC progression and may represent a novel promising target for antimetastatic therapy.
Collapse
Affiliation(s)
- Lingling Xia
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Shi Li
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Yang Liu
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Department of Oncology, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Yuqian Huang
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Beibei Ni
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Lili Wan
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Department of Oncology, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Hongbing Mei
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Xianxin Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Zhiming Cai
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Zesong Li
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
18
|
Many Cells Make Life Work-Multicellularity in Stem Cell-Based Cardiac Disease Modelling. Int J Mol Sci 2018; 19:ijms19113361. [PMID: 30373227 PMCID: PMC6274721 DOI: 10.3390/ijms19113361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cardiac disease causes 33% of deaths worldwide but our knowledge of disease progression is still very limited. In vitro models utilising and combining multiple, differentiated cell types have been used to recapitulate the range of myocardial microenvironments in an effort to delineate the mechanical, humoral, and electrical interactions that modulate the cardiac contractile function in health and the pathogenesis of human disease. However, due to limitations in isolating these cell types and changes in their structure and function in vitro, the field is now focused on the development and use of stem cell-derived cell types, most notably, human-induced pluripotent stem cell-derived CMs (hiPSC-CMs), in modelling the CM function in health and patient-specific diseases, allowing us to build on the findings from studies using animal and adult human CMs. It is becoming increasingly appreciated that communications between cardiomyocytes (CMs), the contractile cell of the heart, and the non-myocyte components of the heart not only regulate cardiac development and maintenance of health and adult CM functions, including the contractile state, but they also regulate remodelling in diseases, which may cause the chronic impairment of the contractile function of the myocardium, ultimately leading to heart failure. Within the myocardium, each CM is surrounded by an intricate network of cell types including endothelial cells, fibroblasts, vascular smooth muscle cells, sympathetic neurons, and resident macrophages, and the extracellular matrix (ECM), forming complex interactions, and models utilizing hiPSC-derived cell types offer a great opportunity to investigate these interactions further. In this review, we outline the historical and current state of disease modelling, focusing on the major milestones in the development of stem cell-derived cell types, and how this technology has contributed to our knowledge about the interactions between CMs and key non-myocyte components of the heart in health and disease, in particular, heart failure. Understanding where we stand in the field will be critical for stem cell-based applications, including the modelling of diseases that have complex multicellular dysfunctions.
Collapse
|
19
|
Jung TW, Park HS, Choi GH, Kim D, Jeong JH, Lee T. Chitinase‐3‐like protein 1 ameliorates atherosclerotic responses via PPARδ‐mediated suppression of inflammation and ER stress. J Cell Biochem 2018; 119:6795-6805. [DOI: 10.1002/jcb.26873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/21/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Tae Woo Jung
- Research Administration TeamSeoul National University Bundang HospitalSeongnamKorea
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Hyung Sub Park
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Geum Hee Choi
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Daehwan Kim
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Ji Hoon Jeong
- Department of PharmacologyCollege of MedicineChung‐Ang UniversitySeoulKorea
| | - Taeseung Lee
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
- Department of SurgerySeoul National University College of MedicineSeoulKorea
| |
Collapse
|
20
|
Wu YS, Zhu B, Luo AL, Yang L, Yang C. The Role of Cardiokines in Heart Diseases: Beneficial or Detrimental? BIOMED RESEARCH INTERNATIONAL 2018; 2018:8207058. [PMID: 29744364 PMCID: PMC5878913 DOI: 10.1155/2018/8207058] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/19/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality, imposing a major disease burden worldwide. Therefore, there is an urgent need to identify new therapeutic targets. Recently, the concept that the heart acts as a secretory organ has attracted increasing attention. Proteins secreted by the heart are called cardiokines, and they play a critical physiological role in maintaining heart homeostasis or responding to myocardial damage and thereby influence the development of heart diseases. Given the critical role of cardiokines in heart disease, they might represent a promising therapeutic target. This review will focus on several cardiokines and discuss their roles in the pathogenesis of heart diseases and as potential therapeutics.
Collapse
Affiliation(s)
- Ye-Shun Wu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Ai-Lin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| |
Collapse
|
21
|
Song HF, He S, Li SH, Yin WJ, Wu J, Guo J, Shao ZB, Zhai XY, Gong H, Lu L, Wei F, Weisel RD, Xie J, Li RK. Aged Human Multipotent Mesenchymal Stromal Cells Can Be Rejuvenated by Neuron-Derived Neurotrophic Factor and Improve Heart Function After Injury. ACTA ACUST UNITED AC 2017; 2:702-716. [PMID: 30062183 PMCID: PMC6059002 DOI: 10.1016/j.jacbts.2017.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 05/02/2017] [Accepted: 07/20/2017] [Indexed: 12/17/2022]
Abstract
The benefits of cell transplantation for cardiac repair are diminished in older individuals and effective methods to rejuvenate aged stem cells are needed to treat the increasing number of older patients with heart failure. Over-expressing NDNF in old hBM-MSCs rejuvenated the cells, increasing their proliferative capacity and reducing cellular apoptosis. In vivo engraftment of NDNF-overexpressing old hBM-MSCs into the ischemic area of mouse hearts improved cardiac function after myocardial infarction, while promoting engrafted stem cell survival and proliferation and decreasing cell senescence. NDNF rejuvenated aged human stem cells, improving their capability to repair the aged heart after ischemic injury through Activation of Akt singling.
Reduced regenerative capacity of aged stem cells hampers the benefits of autologous cell therapy for cardiac regeneration. This study investigated whether neuron-derived neurotrophic factor (NDNF) could rejuvenate aged human bone marrow (hBM)- multipotent mesenchymal stromal cells (MSCs) and whether the rejuvenated hBM-MSCs could improve cardiac repair after ischemic injury. Over-expression of NDNF in old hBM-MSCs decreased cell senescence and apoptosis. Engraftment of NDNF over-expressing old hBM-MSCs into the ischemic area of mouse hearts resulted in improved cardiac function after myocardial infarction, while promoting implanted stem cell survival. Our findings suggest NDNF could be a new factor to rejuvenate aged stem cells and improve their capability to repair the aged heart after injury.
Collapse
Affiliation(s)
- Hui-Fang Song
- Department of Anatomy, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Sheng He
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shu-Hong Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Wen-Juan Yin
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Jun Wu
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Jian Guo
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Zheng-Bo Shao
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
- Department of Ophthalmology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao-Yan Zhai
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Hui Gong
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Li Lu
- Department of Anatomy, Shanxi Medical University, Taiyuan, China
| | - Fang Wei
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Richard D. Weisel
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Jun Xie
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
- Addresses for correspondence: Dr. Jun Xie, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China.
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Canada
- Dr. Ren-Ke Li, Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario M5G 1L7, Canada.
| |
Collapse
|
22
|
Wang L, Yu J, Fordjour PA, Xing X, Gao H, Li Y, Li L, Zhu Y, Gao X, Fan G. Danshen injection prevents heart failure by attenuating post-infarct remodeling. JOURNAL OF ETHNOPHARMACOLOGY 2017; 205:22-32. [PMID: 28465251 DOI: 10.1016/j.jep.2017.04.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/16/2017] [Accepted: 04/26/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danshen Injection (DSI) is a traditional Chinese medicine extracted from Danshen, prepared from the dried root and rhizome of Salvia miltiorrhiza Bunge. Danshen is an ancient antipyretic traditional Chinese medicine which is mostly used to improve blood circulation and dispel blood stasis. Danshen decoction or liquor-fried Danshen (with grain-based liquor) which is cool in nature is traditionally used to 'cool the blood' and reduce the swelling of sores and abscesses. AIM OF STUDY The present study aimed to examine the effect and mechanism of DSI in LAD induced heart injury. MATERIALS AND METHODS One day after LAD surgery, adult male Sprague-Dawley rats were randomized to 3 groups: MI group; DSI group (1.5ml/kg/d, intramuscular); and Valsartan group (10mg/kg/d, intragastric). Echocardiography and hemodynamic measurements (Pressure-Volume loop) were performed to evaluate cardiac function. Pathological methods (Masson, and Sirus red staining) were used to check myocardial fibrosis. Western blotting assay was used to detect the protein expression of MMP-2. RT-PCR was used to detect the gene expression of MMP-9, MPO, iNOS, Bcl-2 and Bax. RESULTS DSI administration to LAD rats resulted in improved cardiac functions, hemodynamic parameters and normalized ventricular mass. Furthermore, DSI-treated group demonstrated potential regulation of myocardial collagen I and III deposition associated with MMP-2 expression. Also, DSI administration decreased gene expression of iNOS, MPO and MMP-9, and increased Bcl-2/Bax ratio. CONCLUSION Myocardial fibrosis, cardiac hypertrophy, hemodynamic deterioration as well as systolic and diastolic dysfunctions which characterize a failing hearts were significantly prevented by DSI. Our study may provide future directions to focus on the anti-hypertrophic mechanisms of DSI and pathological roles played by MMP-2 in myocardial hypertrophy. Meanwhile, DSI also performed the effect of anti-inflammation by the way of decreasing iNOS and MPO. The way Danshen Injection increasing Bcl-2/Bax presented the possibility that it may has the effect of inhibiting cell death.
Collapse
Affiliation(s)
- Lingyan Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Jiahui Yu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Patrick Asare Fordjour
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Xiaoxue Xing
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Hui Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Yanyan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Lingyan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Yan Zhu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China; State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Medicine, Nankai District, Tianjin, PR China.
| | - Guanwei Fan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China; Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, NanKai District, Tianjin, PR China.
| |
Collapse
|
23
|
Liu N, Shi YF, Diao HY, Li YX, Cui Y, Song XJ, Tian X, Li TY, Liu B. MicroRNA-135a Regulates Apoptosis Induced by Hydrogen Peroxide in Rat Cardiomyoblast Cells. Int J Biol Sci 2017; 13:13-21. [PMID: 28123342 PMCID: PMC5264257 DOI: 10.7150/ijbs.16769] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress and apoptosis are the most important pathologic features of ischemic heart disease. Recent research has indicated that microRNAs (miRs) play an essential role in apoptosis. However, whether miRs might regulate B cell lymphoma-2 (Bcl-2) protein in apoptosis during ischemic heart disease is still unclear. The aim of this study, therefore, was to confirm the regulation of microRNA-135a (miR-135a) in oxidative stress injuries induced by hydrogen peroxide (H2O2) in rat cardiomyoblast cells H9c2. To this end, we analyzed the effects of H2O2 treatment on miR-135a expression in rat cardiomyocytes. Furthermore, we upregulated and inhibited miR-135a using mimics and inhibitors, respectively, and examined the effects on cell viability and apoptosis-related proteins. We observed that miR-135a was markedly up-regulated under H2O2 treatment in rat cardiomyoblast cells. Overexpression of miR-135a blocked the Bcl-2 protein and enhanced the apoptosis induced by H2O2, and miR-135a inhibition restored Bcl-2 protein expression. Interestingly, miR-135a inhibition did not attenuate H2O2-induced apoptosis with Bcl-2 knockdown. The results of the present study indicate that miR-135a regulates H2O2-induced apoptosis in H9c2 cells via targeting Bcl-2, and that miR-135a may be a novel therapeutic target for ischemic heart disease.
Collapse
Affiliation(s)
- Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yong-Feng Shi
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Hong-Ying Diao
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yang-Xue Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yan Cui
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Xian-Jing Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Xin Tian
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Tian-Yi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| |
Collapse
|
24
|
Ouchi N, Ohashi K, Shibata R, Murohara T. Protective Roles of Adipocytokines and Myokines in Cardiovascular Disease. Circ J 2016; 80:2073-80. [DOI: 10.1253/circj.cj-16-0663] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Noriyuki Ouchi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine
| | - Koji Ohashi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| |
Collapse
|