1
|
Takada YK, Takada Y. Neuregulin-1 (NRG1) Binds to the Allosteric Binding Site (Site 2) and Suppresses Allosteric Integrin Activation by Inflammatory Cytokines: A Potential Mechanism of Anti-Inflammatory and Anti-Fibrosis Action of NRG1. Cells 2025; 14:617. [PMID: 40277942 PMCID: PMC12025393 DOI: 10.3390/cells14080617] [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: 01/23/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
We showed that multiple inflammatory cytokines (e.g., CCL5, CXCL12, CX3CL1, CD40L, and FGF2) bind to the allosteric site (site 2) of integrins, distinct from the classical RGD-binding site (site 1), and allosterically activate integrins. A major inflammatory lipid mediator 25-hydroxycholesterol is known to bind to site 2 and allosterically activates integrins and induces inflammatory signals (e.g., IL-6 and TNF secretion). Thus, site 2 is involved in inflammatory signaling. Neuregulin-1 (NRG1) is known to suppresses the progression of inflammatory diseases, fibrosis, and insulin resistance. But, the mechanism of anti-inflammatory action of NRG1 is unclear. We previously showed that NRG1 binds to the classical RGD-binding site (site 1). Mutating the 3 Lys residues that are involved in site 1 binding (NRG1 3KE mutant) is defective in binding to site 1 and in ErbB3-mediated mitogenic signals. Docking simulation predicted that NRG1 binds to site 2. We hypothesized that NRG1 acts as an antagonist of site 2 and blocks allosteric activation by multiple cytokines. Here, we describe that NRG1 binds to site 2 but does not activate soluble αvβ3 or αIIbβ3 in 1 mM Ca2+, unlike inflammatory cytokines. Instead, NRG1 suppressed integrin activation by several inflammatory cytokines, suggesting that NRG1 acts as a competitive inhibitor of site 2. Wild-type NRG1 is not suitable for long-term treatment due to its mitogenicity. We showed that the non-mitogenic NRG1 3KE mutant still bound to site 2 and inhibited allosteric activation of soluble and cell-surface integrins, suggesting that NRG1 3KE may have potential as a therapeutic.
Collapse
Affiliation(s)
- Yoko K. Takada
- Department of Dermatology, University of California School of Medicine, Research III Suite 3300, 4645 Second Ave., Sacramento, CA 95817, USA
| | - Yoshikazu Takada
- Department of Dermatology, University of California School of Medicine, Research III Suite 3300, 4645 Second Ave., Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, University of California School of Medicine, Research III Suite 3300, 4645 Second Ave., Sacramento, CA 95817, USA
- VA Northern California Health Care System, 150 Muir Road, Martinez, CA 94553, USA
| |
Collapse
|
2
|
Shi H, Zou Y, Li Y, Li Y, Liu B. Neuregulin-1 reduces Doxorubicin-induced cardiotoxicity by upregulating YAP to inhibit senescence. Int Immunopharmacol 2024; 143:113278. [PMID: 39405937 DOI: 10.1016/j.intimp.2024.113278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024]
Abstract
The cardiotoxicity of Doxorubicin (Dox) limits its clinical application, creating an urgent need to investigate its underlying mechanism and develop effective therapies. Senescence plays an important role in Dox-induced cardiotoxicity (DIC). Recently, Neuregulin-1 (NRG1) was found to regulate Yes-associated protein (YAP), which was reported to inhibit senescence, suggesting that NRG1 might be used to treat DIC by inhibiting senescence through YAP regulation. We examined the changes and regulatory roles of YAP and senescence in Dox cardiotoxicity and whether NRG1 could reduce DIC in chronic DIC mice and Dox-treated H9c2 cells. Our study revealed that sustained small doses of Dox impaired cardiac function and H9c2 cell viability, induced myocardial senescence, and inhibited YAP expression. Conversely, high levels of YAP inhibited Dox-induced senescence in H9c2 cells, indicating that Dox promotes myocardial senescence by inhibiting YAP. In addition, we found that exogenous NRG1 inhibited the phosphorylation of LATS1 and MST1, thereby inhibiting YAP phosphorylation and promote the nuclear translocation of YAP, inhibiting senescence and attenuating Dox-induced cardiotoxicity. YAP knockdown or inhibition of YAP binding to TEA domain transcription factor protein (TEAD)blocks the protective effects of NRG1. In conclusion, our study suggests that Dox-induced myocardial senescence through YAP inhibition is one of the pathological mechanisms of its cardiotoxicity. Additionally, NRG1 reduces DIC by upregulating YAP to inhibit senescence.
Collapse
Affiliation(s)
- Henghe Shi
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yifei Zou
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yinghao Li
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yangxue Li
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China.
| |
Collapse
|
3
|
Ahmad F, Qaisar R. Nicotinamide riboside kinase 2: A unique target for skeletal muscle and cardiometabolic diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167487. [PMID: 39216649 DOI: 10.1016/j.bbadis.2024.167487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Myopathy leads to skeletal and cardiac muscle degeneration which is a major cause of physical disability and heart failure. Despite the therapeutic advancement the prevalence of particularly cardiac diseases is rising at an alarming rate and novel therapeutic targets are required. Nicotinamide riboside kinase-2 (NRK-2 or NMRK2) is a muscle-specific β1-integrin binding protein abundantly expressed in the skeletal muscle while only a trace amount is detected in the healthy cardiac muscle. The level in cardiac tissue is profoundly upregulated under pathogenic conditions such as ischemia and hypertension. NRK-2 was initially identified to regulate myoblast differentiation and to enhance the levels of NAD+, an important coenzyme that potentiates cellular energy production and stress resilience. Recent advancement has shown that NRK-2 critically regulates numerous cellular and molecular processes under pathogenic conditions to modulate the disease severity. Therefore, given its restricted expression in the cardiac and skeletal muscle, NRK-2 may serve as a unique therapeutic target. In this review, we provided a comprehensive overview of the diverse roles of NRK-2 played in different cardiac and muscular diseases and discussed the underlying molecular mechanisms in detail. Moreover, this review precisely examined how NRK-2 regulates metabolism in cardiac muscle, and how dysfunctional NRK-2 is associated with energetic deficit and impaired muscle function, manifesting various cardiac and skeletal muscle disease conditions.
Collapse
Affiliation(s)
- Firdos Ahmad
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rizwan Qaisar
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
4
|
Ahmad F, Gupta A, Marzook H, Woodgett JR, Saleh MA, Qaisar R. Natural compound screening predicts novel GSK-3 isoform-specific inhibitors. Biochimie 2024; 225:68-80. [PMID: 38723940 DOI: 10.1016/j.biochi.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/21/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024]
Abstract
Glycogen synthase kinase-3 (GSK-3) plays important roles in the pathogenesis of cardiovascular, metabolic, neurological disorders and cancer. Isoform-specific loss of either GSK-3α or GSK-3β often provides cytoprotective effects under such clinical conditions. However, available synthetic small molecule inhibitors are relatively non-specific, and their chronic use may lead to adverse effects. Therefore, screening for natural compound inhibitors to identify the isoform-specific inhibitors may provide improved clinical utility. Here, we screened 70 natural compounds to identify novel natural GSK-3 inhibitors employing comprehensive in silico and biochemical approaches. Molecular docking and pharmacokinetics analysis identified two natural compounds Psoralidin and Rosmarinic acid as potential GSK-3 inhibitors. Specifically, Psoralidin and Rosmarinic acid exhibited the highest binding affinities for GSK-3α and GSK-3β, respectively. Consistent with in silico findings, the kinase assay-driven IC50 revealed superior inhibitory effects of Psoralidin against GSK-3α (IC50 = 2.26 μM) vs. GSK-3β (IC50 = 4.23 μM) while Rosmarinic acid was found to be more potent against GSK-3β (IC50 = 2.24 μM) than GSK-3α (IC50 = 5.14 μM). Taken together, these studies show that the identified natural compounds may serve as GSK-3 inhibitors with Psoralidin serving as a better inhibitor for GSK-3α and Rosmarinic for GSK-3β isoform, respectively. Further characterization employing in vitro and preclinical models will be required to test the utility of these compounds as GSK-3 inhibitors for cardiometabolic and neurological disorders and cancers.
Collapse
Affiliation(s)
- Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Space Medicine Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| | - Anamika Gupta
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hezlin Marzook
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mohamed A Saleh
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Space Medicine Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
5
|
Erciyes D, Bora ES, Tekindal MA, Erbaş O. Demonstration of the Protective Effect of Vinpocetine in Diabetic Cardiomyopathy. J Clin Med 2024; 13:4637. [PMID: 39200779 PMCID: PMC11354616 DOI: 10.3390/jcm13164637] [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: 07/11/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Diabetic cardiomyopathy (DCM) poses a significant risk for heart failure in individuals with diabetes, yet its underlying mechanisms remain incompletely understood. Elevated blood sugar levels initiate harmful processes, including apoptosis, collagen accumulation, and fibrosis in the heart. Vinpocetine, a derivative of Vinca minor L., has demonstrated diverse pharmacological effects, including vasodilation, anti-inflammatory properties, and enhanced cellular metabolism. This study aims to investigate Vinpocetine's protective and remodeling effects in diabetic cardiomyopathy by evaluating biochemical and histopathological parameters. Methods: Twenty-one adult male Wistar rats were induced with diabetes using streptozocin and divided into Diabetes and Diabetes + Vinpocetine groups. Histopathological analyses, TGF-β1 immunoexpression, and measurements of plasma markers (TGF-β, pro-BNP, Troponin T) were performed. Biochemical analyses included HIF-1 alpha and neuregulin-1β quantification and evaluation of lipid peroxidation. Results: Vinpocetine significantly reduced cardiac muscle thickness, TGF-β1 expression, and plasma in diabetic rats. HIF-1 alpha and neuregulin-1β levels increased with Vinpocetine treatment. Histopathological observations confirmed reduced fibrosis and structural abnormalities in Vinpocetine-treated hearts. Conclusions: This study provides comprehensive evidence supporting the protective effects of Vinpocetine against diabetic cardiomyopathy. Vinpocetine treatment improved cardiac morphology, immunohistochemistry, and modulation of biochemical markers, suggesting its potential as a therapeutic intervention to attenuate the negative impact of diabetes on heart function.
Collapse
Affiliation(s)
- Demet Erciyes
- Department of Cardiology, Faculty of Medicine, Demiroğlu Bilim University, 34394 Istanbul, Türkiye;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, 35620 Izmir, Türkiye
| | - Mustafa Agah Tekindal
- Department of Basic Medical Sciences Biostatistics, Faculty of Medicine, İzmir Katip Çelebi Unıversity, 35620 Izmir, Türkiye;
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, 34394 Istanbul, Türkiye;
| |
Collapse
|
6
|
Zhang W, Dao JJ, Li Q, Liu C, Qiao CM, Cui C, Shen YQ, Zhao WJ. Neuregulin 1 mitigated prolactin deficiency through enhancing TRPM8 signaling under the influence of melatonin in senescent pituitary lactotrophs. Int J Biol Macromol 2024; 275:133659. [PMID: 38969045 DOI: 10.1016/j.ijbiomac.2024.133659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
The age-related alterations in pituitary function, including changes in prolactin (PRL) production contributes to the systemic susceptibility to age-related diseases. Our previous research has shown the involvement of Nrg1 in regulating the expression and secretion of PRL. However, the precise role of Nrg1 in mitigating the senescence of pituitary lactotrophs and the underlying mechanisms are yet to be comprehended. Here, data from the GEPIA database was used to evaluate the association between transient receptor potential cation channel subfamily M member 8 (TRPM8) and PRL in normal human pituitary tissues, followed by immunofluorescence verification using a human pituitary tissue microarray. TRPM8 levels showed a significant positive association with PRL expression in normal human pituitary tissues, and both TRPM8 and PRL levels declined during aging, suggesting that TRPM8 may regulate pituitary aging by affecting PRL production. It was also found that treatment with exogenous neuregulin 1 (Nrg1) markedly delayed the senescence of GH3 cells (rat lactotroph cell line) generated by D-galactose (D-gal). In addition, melatonin reduced the levels of senescence-related markers in senescent pituitary cells by promoting Nrg1 / ErbB4 signaling, stimulating PRL expression and secretion. Further investigation showed that Nrg1 attenuated senescence in pituitary cells by increasing TRPM8 expression. Downregulation of TRPM8 activation eliminated Nrg1-mediated amelioration of pituitary cell senescence. These findings demonstrate the critical function of Nrg1 / ErbB signaling in delaying pituitary lactotroph cell senescence and enhancing PRL production via promoting TRPM8 expression under the modulation of melatonin.
Collapse
Affiliation(s)
- Wei Zhang
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China; Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang 550000, Guizhou, China
| | - Ji-Ji Dao
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Qian Li
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Chong Liu
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Chen-Meng Qiao
- Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Chun Cui
- Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Yan-Qin Shen
- Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Wei-Jiang Zhao
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China.
| |
Collapse
|
7
|
Xu Q, Xiao Z, Yang Q, Yu T, Deng X, Chen N, Huang Y, Wang L, Guo J, Wang J. Hydrogel-based cardiac repair and regeneration function in the treatment of myocardial infarction. Mater Today Bio 2024; 25:100978. [PMID: 38434571 PMCID: PMC10907859 DOI: 10.1016/j.mtbio.2024.100978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
A life-threatening illness that poses a serious threat to human health is myocardial infarction. It may result in a significant number of myocardial cells dying, dilated left ventricles, dysfunctional heart function, and ultimately cardiac failure. Based on the development of emerging biomaterials and the lack of clinical treatment methods and cardiac donors for myocardial infarction, hydrogels with good compatibility have been gradually applied to the treatment of myocardial infarction. Specifically, based on the three processes of pathophysiology of myocardial infarction, we summarized various types of hydrogels designed for myocardial tissue engineering in recent years, including natural hydrogels, intelligent hydrogels, growth factors, stem cells, and microRNA-loaded hydrogels. In addition, we also describe the heart patch and preparation techniques that promote the repair of MI heart function. Although most of these hydrogels are still in the preclinical research stage and lack of clinical trials, they have great potential for further application in the future. It is expected that this review will improve our knowledge of and offer fresh approaches to treating myocardial infarction.
Collapse
Affiliation(s)
- Qiaxin Xu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, 510630, China
| | - Qianzhi Yang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Tingting Yu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Xiujiao Deng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Nenghua Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Lihong Wang
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Endocrinology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jun Guo
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
8
|
Huang J, Pang X, Zhang X, Qiu W, Zhang X, Wang R, Xie W, Bai Y, Zhou S, Liao J, Xiong Z, Tang Z, Su R. N-acetylcysteine combined with insulin attenuates myocardial injury in canines with type 1 diabetes mellitus by modulating TNF-α-mediated apoptotic pathways and affecting linear ubiquitination. Transl Res 2023; 262:1-11. [PMID: 37422055 DOI: 10.1016/j.trsl.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/24/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
The exact pathogenesis of type 1 diabetes mellitus (DM) is still unclear. Numerous organs, including the heart, will suffer damage and malfunction as a result of long-term hyperglycemia. Currently, insulin therapy alone is still not the best treatment for type 1 DM. In order to properly treat and manage patients with type 1 DM, it is vital to seek a combination that includes both insulin and additional medications. This study aims to explore the therapeutic effect and mechanism of N-acetylcysteine (NAC) combined with insulin on type 1 DM. By giving beagle canines injections of streptozotocin (STZ) and alloxan (ALX) (20 mg/kg each), a model of type 1 DM was created. The results showed that this combination could effectively control blood sugar level, improve heart function, avoid the damage of mitochondria and myocardial cells, and prevent the excessive apoptosis of myocardial cells. Importantly, the combination can activate nuclear factor kappa-B (NF-κB) by promoting linear ubiquitination of receptor-interacting protein kinase 1 (RIPK1) and NF-κB-essential modulator (NEMO) and inhibitor of NF-κB (IκB) phosphorylation. The combination can increase the transcription and linear ubiquitination of Cellular FLICE (FADD-like IL-1β-converting enzyme) -inhibitory protein (c-FLIP), diminish the production of cleaved-caspase-8 p18 and cleaved-caspase-3 to reduce apoptosis. This study confirmed that NAC combined with insulin can promote the linear ubiquitination of RIPK1, NEMO and c-FLIP and regulate the apoptosis pathway mediated by TNF-α to attenuate the myocardial injury caused by type 1 DM. Meanwhile, the research served as a resource when choosing a clinical strategy for DM cardiac complications.
Collapse
Affiliation(s)
- Jianjia Huang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiaoyue Pang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xinting Zhang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wenyue Qiu
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xuluan Zhang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Rongmei Wang
- Department of Animal Science, Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, Guangdong, China
| | - Wenting Xie
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yuman Bai
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shuilian Zhou
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jianzhao Liao
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhaojun Xiong
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaoxin Tang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Rongsheng Su
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Ahmad F, Marzook H, Gupta A, Aref A, Patil K, Khan AA, Saleh MA, Koch WJ, Woodgett JR, Qaisar R. GSK-3α aggravates inflammation, metabolic derangement, and cardiac injury post-ischemia/reperfusion. J Mol Med (Berl) 2023; 101:1379-1396. [PMID: 37707557 DOI: 10.1007/s00109-023-02373-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/18/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Reperfusion after acute myocardial infarction further exaggerates cardiac injury and adverse remodeling. Irrespective of cardiac cell types, loss of specifically the α isoform of the protein kinase GSK-3 is protective in chronic cardiac diseases. However, the role of GSK-3α in clinically relevant ischemia/reperfusion (I/R)-induced cardiac injury is unknown. Here, we challenged cardiomyocyte-specific conditional GSK-3α knockout (cKO) and littermate control mice with I/R injury and investigated the underlying molecular mechanism using an in vitro GSK-3α gain-of-function model in AC16 cardiomyocytes post-hypoxia/reoxygenation (H/R). Analysis revealed a significantly lower percentage of infarct area in the cKO vs. control hearts post-I/R. Consistent with in vivo findings, GSK-3α overexpression promoted AC16 cardiomyocyte death post-H/R which was accompanied by an induction of reactive oxygen species (ROS) generation. Consistently, GSK-3α gain-of-function caused mitochondrial dysfunction by significantly suppressing mitochondrial membrane potential. Transcriptomic analysis of GSK-3α overexpressing cardiomyocytes challenged with hypoxia or H/R revealed that NOD-like receptor (NLR), TNF, NF-κB, IL-17, and mitogen-activated protein kinase (MAPK) signaling pathways were among the most upregulated pathways. Glutathione and fatty acid metabolism were among the top downregulated pathways post-H/R. Together, these observations suggest that loss of cardiomyocyte-GSK-3α attenuates cardiac injury post-I/R potentially through limiting the myocardial inflammation, mitochondrial dysfunction, and metabolic derangement. Therefore, selective inhibition of GSK-3α may provide beneficial effects in I/R-induced cardiac injury and remodeling. KEY MESSAGES: GSK-3α promotes cardiac injury post-ischemia/reperfusion (I/R). GSK-3α regulates inflammatory and metabolic pathways post-hypoxia/reoxygenation (H/R). GSK-3α overexpression upregulates NOD-like receptor (NLR), TNF, NF-kB, IL-17, and MAPK signaling pathways in cardiomyocytes post-H/R. GSK-3α downregulates glutathione and fatty acid metabolic pathways in cardiomyocytes post-H/R.
Collapse
Affiliation(s)
- Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE.
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE.
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240, USA.
| | - Hezlin Marzook
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Anamika Gupta
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Aseel Aref
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Kiran Patil
- LifeBytes India Pvt Ltd., Brigade Triumph, Hebbal Kempapura, Bengaluru, Karnataka, 560092, India
| | - Amir Ali Khan
- Department of Applied Biology, College of Science, University of Sharjah, Sharjah, 27272, UAE
- BioGrad Biobank, 61 Stephenson Way, Liverpool, L13 1HN, UK
| | - Mohamed A Saleh
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Walter J Koch
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| |
Collapse
|
10
|
Ahmad F, Karim A, Khan J, Qaisar R. Circulating H-FABP as a biomarker of frailty in patients with chronic heart failure. Exp Biol Med (Maywood) 2023; 248:1383-1392. [PMID: 37787063 PMCID: PMC10657591 DOI: 10.1177/15353702231198080] [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: 04/14/2023] [Accepted: 07/10/2023] [Indexed: 10/04/2023] Open
Abstract
Increased vulnerability to physiologic stressors, termed frailty, is a common occurrence in patients with chronic heart failure (CHF). However, the definite biomarkers to assess frailty in CHF patients are not known. Here, we assessed the frailty phenotype and its potential association with heart failure (HF) markers in CHF patients. We categorized controls (n = 59) and CHF patients (n = 80), the participants, into robust, pre-frail, and frail based on the cardiovascular health study (CHS) frailty index. The plasma levels of HF markers, including tumorigenicity 2 (s-ST2), galectin-3, and heart-type fatty acid binding protein (H-FABP), were measured and correlated with frailty phenotype and cardiac function. The levels of plasma s-ST2, galectin-3, and H-FABP were profoundly elevated in CHF patients. Conversely, the frailty index scores were significantly lower in ischemic and non-ischemic CHF patients versus controls. Of the assessed HF markers, only H-FABP was positively correlated (r2 = 0.07, P = 0.02) with the frailty score in CHF patients. Collectively, these observations suggest that circulating H-FABP may serve as a biomarker of frailty in CHF patients.
Collapse
Affiliation(s)
- Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Javaidullah Khan
- Department of Cardiology, Post Graduate Medical Institute, Hayatabad Medical Complex, Peshawar 25120, Pakistan
| | - Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
11
|
Marzook H, Gupta A, Tomar D, Saleh MA, Patil K, Semreen MH, Hamoudi R, Soares NC, Qaisar R, Ahmad F. Nicotinamide riboside kinase-2 regulates metabolic adaptation in the ischemic heart. J Mol Med (Berl) 2023; 101:311-326. [PMID: 36808555 DOI: 10.1007/s00109-023-02296-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/18/2023] [Accepted: 02/06/2023] [Indexed: 02/23/2023]
Abstract
Ischemia-induced metabolic remodeling plays a critical role in the pathogenesis of adverse cardiac remodeling and heart failure however, the underlying molecular mechanism is largely unknown. Here, we assess the potential roles of nicotinamide riboside kinase-2 (NRK-2), a muscle-specific protein, in ischemia-induced metabolic switch and heart failure through employing transcriptomic and metabolomic approaches in ischemic NRK-2 knockout mice. The investigations revealed NRK-2 as a novel regulator of several metabolic processes in the ischemic heart. Cardiac metabolism and mitochondrial function and fibrosis were identified as top dysregulated cellular processes in the KO hearts post-MI. Several genes linked to mitochondrial function, metabolism, and cardiomyocyte structural proteins were severely downregulated in the ischemic NRK-2 KO hearts. Analysis revealed significantly upregulated ECM-related pathways which was accompanied by the upregulation of several key cell signaling pathways including SMAD, MAPK, cGMP, integrin, and Akt in the KO heart post-MI. Metabolomic studies identified profound upregulation of metabolites mevalonic acid, 3,4-dihydroxyphenylglycol, 2-penylbutyric acid, and uridine. However, other metabolites stearic acid, 8,11,14-eicosatrienoic acid, and 2-pyrrolidinone were significantly downregulated in the ischemic KO hearts. Taken together, these findings suggest that NRK-2 promotes metabolic adaptation in the ischemic heart. The aberrant metabolism in the ischemic NRK-2 KO heart is largely driven by dysregulated cGMP and Akt and mitochondrial pathways. KEY MESSAGES: Post-myocardial infarction metabolic switch critically regulates the pathogenesis of adverse cardiac remodeling and heart failure. Here, we report NRK-2 as a novel regulator of several cellular processes including metabolism and mitochondrial function post-MI. NRK-2 deficiency leads to downregulation of genes important for mitochondrial pathway, metabolism, and cardiomyocyte structural proteins in the ischemic heart. It was accompanied by upregulation of several key cell signaling pathways including SMAD, MAPK, cGMP, integrin, and Akt and dysregulation of numerous metabolites essential for cardiac bioenergetics. Taken together, these findings suggest that NRK-2 is critical for metabolic adaptation of the ischemic heart.
Collapse
Affiliation(s)
- Hezlin Marzook
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
| | - Anamika Gupta
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
| | - Dhanendra Tomar
- Department of Internal Medicine, Section On Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mohamed A Saleh
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Kiran Patil
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
| | - Mohammad H Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. 27272, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Division of Surgery and Interventional Science, University College London, London, W1W 7EJ, UK
| | - Nelson C Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. 27272, Sharjah, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av.a Padre Cruz, Lisbon, 1649-016, Portugal
| | - Rizwan Qaisar
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Firdos Ahmad
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates.
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, 59911, Abu Dhabi, United Arab Emirates.
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240, USA.
| |
Collapse
|
12
|
Ahmad F, Karim A, Khan J, Qaisar R. Plasma Galectin-3 and H-FABP correlate with poor physical performance in patients with congestive heart failure. Exp Biol Med (Maywood) 2023; 248:532-540. [PMID: 36803120 PMCID: PMC10281532 DOI: 10.1177/15353702231151980] [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: 10/18/2022] [Accepted: 12/26/2022] [Indexed: 02/22/2023] Open
Abstract
Heart failure (HF) is often associated with compromised physical capacity in patients. However, it is unclear if established HF markers correlate with the physical performance of patients with congestive HF (CHF). We assessed the left ventricular end-systolic dimension (LVESD) and ejection fraction (LVEF) and, physical performance parameters, including short physical performance battery (SPPB), gait speed (GS), and handgrip strength (HGS) in 80 patients with CHF along with 59 healthy controls. Furthermore, levels of plasma HF markers galectin-3 and heart-specific fatty acid binding protein (H-FABP) were measured in relation to the severity of HF and physical performance. Irrespective of etiology, significantly greater LVESD and lower LVEF were observed in HF patients versus controls. As expected, the levels of HF markers galectin-3 and H-FABP were upregulated in the CHF patients which were accompanied by significantly elevated levels of plasma zonulin and inflammatory marker C-reactive protein (CRP). The SPPB scores, GS, and HGS were significantly lower in the ischemic and non-ischemic HF patients than controls. The level of galectin-3 was inversely correlated with SPPB scores (r2 = 0.089, P = 0.01) and HGS (r2 = 0.078, P = 0.01). Similarly, H-FABP levels were also inversely correlated with SPPB scores (r2 = 0.06, P = 0.03) and HGS (r2 = 0.109, P = 0.004) in the patients with CHF. Taken together, CHF adversely affects physical performance, and galectin-3 and H-FABP may serve as biomarkers of physical disability in patients with CHF. The robust correlations of galectin-3 and H-FABP with the physical performance parameters and CRP in CHF patients suggest that the poor physical performance may partly be caused due to systemic inflammation.
Collapse
Affiliation(s)
- Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, 59911 United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Javaidullah Khan
- Department of Cardiology, Post Graduate Medical Institute, Hayatabad Medical Complex, Peshawar 25120, Pakistan
| | - Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
13
|
Chang T, Liu C, Yang H, Lu K, Han Y, Zheng Y, Huang H, Wu Y, Song Y, Yu Q, Shen Z, Jiang T, Zhang Y. Fibrin-based cardiac patch containing neuregulin-1 for heart repair after myocardial infarction. Colloids Surf B Biointerfaces 2022; 220:112936. [DOI: 10.1016/j.colsurfb.2022.112936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/11/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022]
|
14
|
Zhbanov KA, Salakheeva EY, Sokolova IY, Zheleznykh EA, Zektser VY, Privalova EV, Belenkov YN, Shchendrygina AA. Neuregulin-1β, Biomarkers of Inflammation and Myocardial Fibrosis in Heart Failure Patients. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-09-05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- K. A. Zhbanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. Yu. Salakheeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. Ya. Sokolova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. A. Zheleznykh
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - V. Yu. Zektser
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. V. Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - Yu. N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | |
Collapse
|
15
|
Wang C, Qin Y, Zhang X, Yang Y, Wu X, Liu J, Qin S, Chen K, Xiao W. Effect of Dapagliflozin on Indicators of Myocardial Fibrosis and Levels of Inflammatory Factors in Heart Failure Patients. DISEASE MARKERS 2022; 2022:5834218. [PMID: 36105253 PMCID: PMC9467782 DOI: 10.1155/2022/5834218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022]
Abstract
Objective To explore the effect of dapagliflozin on the myocardial fibrosis and the levels of inflammatory factors in heart failure patients. Methods 60 patients with T2DM who were diagnosed as acute left heart failure or acute exacerbation of chronic left heart failure in the Department of Cardiology of our hospital from November 1, 2020, to December 31, 2021, during hospitalization were the study subjects. According to the treatment regimen, they were divided into the experimental group (EG) which received dapagliflozin and conventional drugs and the control group (CG) which received conventional drugs, with 30 cases in each group to compare and analyze the clinical indicators such as myocardial fibrosis and inflammatory factors. Results The levels of TNF-α, IL-1β, IL-6, and hs-CRP in the two groups were decreased gradually after treatment, and the levels of TNF-α, IL-1β, IL-6, and hs-CRP in the EG were visibly lower compared with those in the CG at week 4 of treatment (P < 0.05). The cardiac function evaluation of patients showed that the levels of LVEF and LVEDD in both groups were gradually improved after treatment, with a significant difference from the fourth week. In other words, compared with the CG, the LVEF level in the EG was obviously higher (P < 0.05), the LVEDD level was distinctly lower (P < 0.05), and the levels of ST2, BNP, and MCP-1 in the EG were clearly lower at week 4 of treatment (P < 0.05) with a statistical significance in difference. Conclusion Dapagliflozin has a definite curative effect in heart failure patients with type 2 diabetes mellitus, which can effectively reduce the inflammatory response of patients and inhibit the myocardial fibrosis, and has a potential value in improving cardiac function and promoting prognosis.
Collapse
Affiliation(s)
- Chuanqiang Wang
- Department of Cardiology, The Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| | - Yiteng Qin
- Basic Medicine College, Hebei Medical University, 050017 Shijiazhuang City, Hebei Province, China
| | - Xiaojun Zhang
- Department of Cardiology, The Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| | - Yang Yang
- Department of Cardiology, The Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| | - Xuan Wu
- Department of Cardiology, The Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| | - Jing Liu
- Department of Cardiology, The Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| | - Shuhui Qin
- Hebei Orthopedic Research Institution, 050051 Shijiazhuang City, Hebei Province, China
| | - Ke Chen
- Catheter Room, Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| | - Wenliang Xiao
- Department of Cardiology, The Third Hospital of Hebei Medical University, 050051 Shijiazhuang City, Hebei Province, China
| |
Collapse
|
16
|
Wang Y, Wei J, Zhang P, Zhang X, Wang Y, Chen W, Zhao Y, Cui X. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022; 13:945206. [PMID: 36120374 PMCID: PMC9471952 DOI: 10.3389/fphar.2022.945206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
NRG1 (Neuregulin-1) is an effective cardiomyocyte proliferator, secreted and released by endothelial vascular cells, and affects the cardiovascular system. It plays a major role in heart growth, proliferation, differentiation, apoptosis, and other cardiovascular processes. Numerous experiments have shown that NRG1 can repair the heart in the pathophysiology of atherosclerosis, myocardial infarction, ischemia reperfusion, heart failure, cardiomyopathy and other cardiovascular diseases. NRG1 can connect related signaling pathways through the NRG1/ErbB pathway, which form signal cascades to improve the myocardial microenvironment, such as regulating cardiac inflammation, oxidative stress, necrotic apoptosis. Here, we summarize recent research advances on the molecular mechanisms of NRG1, elucidate the contribution of NRG1 to cardiovascular disease, discuss therapeutic approaches targeting NRG1 associated with cardiovascular disease, and highlight areas for future research.
Collapse
Affiliation(s)
- Yan Wang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianliang Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peng Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifei Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanan Zhao
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| |
Collapse
|
17
|
Decreased expression of ErbB2 on left ventricular epicardial cells in patients with diabetes mellitus. Cell Signal 2022; 96:110360. [PMID: 35609807 PMCID: PMC9671200 DOI: 10.1016/j.cellsig.2022.110360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/09/2022] [Accepted: 05/18/2022] [Indexed: 11/21/2022]
Abstract
We investigated the cell surface expression of ErbB receptors on left ventricular (LV) epicardial endothelial cells and CD105+ cells obtained from cardiac biopsies of patients undergoing coronary artery bypass grafting surgery (CABG). Endothelial cells and CD105+ non-endothelial cells were freshly isolated from LV epicardial biopsies obtained from 15 subjects with diabetes mellitus (DM) and 8 controls. The expression of ErbB receptors was examined using flow cytometry. We found that diabetes mellitus (DM) and high levels of hemoglobin A1C are associated with reduced expression of ErbB2. To determine if the expression of ErbB2 receptors is regulated by glucose levels, we examined the effect of high Glucose in human microvascular endothelial cells (HMEC-1) and CD105+ non-endothelial cells, using a novel flow cytometric approach to simultaneously determine the total level, cell surface expression, and phosphorylation of ErbB2. Incubation of cells in the presence of 25 mM d-glucose resulted in decreased cell surface but not total levels of ErbB2. The level of ErbB2 at the cell surface is controlled by disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) that is expressed on LV epicardial cells. Inhibition of ADAM10 prevented the high glucose-dependent decrease in the cell surface expression of ErbB2. We suggest that high Glucose depresses ErbB receptor signaling in endothelial cells and cardiac progenitor cells via the promotion of ADAM10-dependent cleavage of ErbB2 at the cell surface, thus contributing to vascular dysfunction and adverse remodeling seen in diabetic patients.
Collapse
|
18
|
Li L, Shang L, Kang W, Lingqian D, Ge S. Neuregulin‐1 promotes the proliferation, migration and angiogenesis of human periodontal ligament stem cells
in vitro. Cell Biol Int 2022; 46:792-805. [PMID: 35077607 DOI: 10.1002/cbin.11770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Ling Li
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
- Department of StomatologyLinyi People's HospitalLinyiShandong ProvinceChina
| | - Lingling Shang
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
| | - Wenyan Kang
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
| | - Du Lingqian
- Department of StomatologyThe Second Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandong ProvinceChina
| | - Shaohua Ge
- Department of PeriodontologySchool and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration
| |
Collapse
|
19
|
Shchendrigina AA, Zhbanov KA, Privalova EV, Iusupova AO, Bytdaeva AH, Danilogorskaya YA, Zheleznykh EA, Suvorov AY, Zektser VY, Mnatsakanyan MG, Lyapidevskaya OV, Khabarova NV, Naymann YI, Belenkov YN, Starostina ES. [Circulating Neuregulin-1 and Chronic Heart Failure with Preserved Ejection]. ACTA ACUST UNITED AC 2020; 60:1222. [PMID: 33487159 DOI: 10.18087/cardio.2020.11.n1222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/08/2020] [Indexed: 11/18/2022]
Abstract
Chronic heart failure (CHF) with preserved ejection fraction (CHFpEF) is an unsolved, socially relevant challenge since it is associated with a high level of morbidity and mortality. Early markers for this pathology are unavailable, and therapeutic approaches are undeveloped. This necessitates extensive studying the mechanisms of CHFpEF to identify therapeutic targets. According to current notions, systemic inflammation and endothelial dysfunction play an important role in the pathogenesis of CHFpEF. These processes induce the development of myocardial fibrosis and impairment of cardiomyocyte relaxation, thereby resulting in diastolic dysfunction and increased left ventricular (LV) filling pressure. Neuregulin-1 (NRG-1) is a paracrine growth factor and a natural agonist of ErbB receptor family synthesized in the endothelium of coronary microvessels. The NRG-1 / ErbB4 system of the heart is activated at early stages of CHFpEF to enhance the cardiomyocyte resistance to oxidative stress. Preclinical and clinical (phases II and III) studies have shown that the recombinant NRG-1 therapy results in improvement of myocardial contractility and in LV reverse remodeling. Results of recent studies suggest possible anti-inflammatory and antifibrotic effects of NRG-1, which warrants studying the activity of this system in patients with CHFpEF.
Collapse
Affiliation(s)
- A A Shchendrigina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - K A Zhbanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E V Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - A O Iusupova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - A H Bytdaeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu A Danilogorskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E A Zheleznykh
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | | | - V Yu Zektser
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - M G Mnatsakanyan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - O V Lyapidevskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - N V Khabarova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu I Naymann
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu N Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | | |
Collapse
|
20
|
Cacciapuoti M, Johnson B, Kapdia A, Powell S, Gallicano GI. The Role of Neuregulin and Stem Cells as Therapy Post-Myocardial Infarction. Stem Cells Dev 2020; 29:1266-1274. [PMID: 32731805 DOI: 10.1089/scd.2020.0099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease, including myocardial infarction (MI), is a leading cause of morbidity and mortality in the United States. Due to the limited self-renewal capacity of cardiac tissue, MIs can lead to progressive heart disease with a lasting impact on health and quality of life. The recent discovery of cardiac stem cells has incited research into their potential therapeutic applications for patients suffering from cardiovascular disease. Studies have demonstrated the ability of stem cells to both generate cardiac tissues in vitro and aid in the recovery of cardiovascular function in vivo in animal models. However, the long-term efficacy of stem cells as regenerative therapy is still unknown. Exploration of alternative therapies is underway, including the use of cardiac growth factor neuregulin-1 (NRG-1). Research has demonstrated that NRG-1 not only has direct effects on cardiomyocytes (CM) but also acts within the tissues supporting the CM. Transplantation of NRG-1 into ischemic cardiac tissue mitigates the progression of heart failure and can reverse cardiac remodeling. Recent publications have sought to study the combined use of these agents, and while the results are promising, they do warrant further research. This review aims to consider these therapies separately as well as in combination.
Collapse
Affiliation(s)
- Maria Cacciapuoti
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Bria Johnson
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Anjani Kapdia
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Sarah Powell
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - G Ian Gallicano
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| |
Collapse
|
21
|
Gong J, Zhou F, Wang SXX, Xu J, Xiao F. Caveolin-3 protects diabetic hearts from acute myocardial infarction/reperfusion injury through β2AR, cAMP/PKA, and BDNF/TrkB signaling pathways. Aging (Albany NY) 2020; 12:14300-14313. [PMID: 32692723 PMCID: PMC7425465 DOI: 10.18632/aging.103469] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
Abstract
Diabetes mellitus (DM) might increase the incidence and mortality of cardiac failure after acute myocardial infarction (AMI) in patients. We attempted to investigate whether Caveolin-3 showed beneficial effects in DM patient post-MI injury through the cAMP/PKA and BDNF/TrkB signaling pathways. The activity of ADRB2 and cAMP/PKA signaling were impaired in nondiabetic ischemia-reperfusion (I/R) group compared with the sham and DM groups and were more impaired in diabetic I/R group than in the I/R group. In H9C2 cells, high-glucose (HG) stimulation further enhanced H/R injury by promoting cell apoptosis, inhibiting cell viability, and suppressing TrkB and Akt signaling; in contrast, the ADRB2 agonist isoprenaline (ISO) significantly attenuated the above-described effects of HG stimulation. Caveolin-3 overexpression promoted the localization of ADRB2 on the membrane of the HG-stimulated H9C2 cells, subsequently inhibiting apoptosis and promoting cell viability. Under HG stimulation, Caveolin-3 overexpression enhanced the activity of the cAMP/PKA and BDNF/TrkB signaling pathways, whereas ADRB2 silencing reversed the effects of Caveolin-3 overexpression. In conclusion, ADRB2 agonist promoted the activity of the BDNF/TrkB and cAMP/PKA signaling pathways, mitigating the HG-aggravated H/R injuries in H9C2 cells. Caveolin-3 exerts a protective effect on diabetic hearts against I/R damage through the β2AR, cAMP/PKA, and BDNF/TrkB signaling pathways.
Collapse
Affiliation(s)
- Jiaji Gong
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Fan Zhou
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Simin Xie Xin Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junmei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feng Xiao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
22
|
Singh AP, Umbarkar P, Guo Y, Force T, Gupte M, Lal H. Inhibition of GSK-3 to induce cardiomyocyte proliferation: a recipe for in situ cardiac regeneration. Cardiovasc Res 2020; 115:20-30. [PMID: 30321309 DOI: 10.1093/cvr/cvy255] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/09/2018] [Indexed: 01/03/2023] Open
Abstract
With an estimated 38 million current patients, heart failure (HF) is a leading cause of morbidity and mortality worldwide. Although the aetiology differs, HF is largely a disease of cardiomyocyte (CM) death or dysfunction. Due to the famously limited amount of regenerative capacity of the myocardium, the only viable option for advanced HF patients is cardiac transplantation; however, donor's hearts are in very short supply. Thus, novel regenerative strategies are urgently needed to reconstitute the injured hearts. Emerging data from our lab and others have elucidated that CM-specific deletion of glycogen synthase kinase (GSK)-3 family of kinases induces CM proliferation, and the degree of proliferation is amplified in the setting of cardiac stress. If this proliferation is sufficiently robust, one could induce meaningful regeneration without the need for delivering exogenous cells to the injured myocardium (i.e. cardiac regeneration in situ). Herein, we will discuss the emerging role of the GSK-3s in CM proliferation and differentiation, including their potential implications in cardiac regeneration. The underlying molecular interactions and cross-talk among signalling pathways will be discussed. We will also review the specificity and limitations of the available small molecule inhibitors targeting GSK-3 and their potential applications to stimulate the endogenous cardiac regenerative responses to repair the injured heart.
Collapse
Affiliation(s)
- Anand Prakash Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Prachi Umbarkar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Yuanjun Guo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| |
Collapse
|
23
|
De Keulenaer GW, Feyen E, Dugaucquier L, Shakeri H, Shchendrygina A, Belenkov YN, Brink M, Vermeulen Z, Segers VFM. Mechanisms of the Multitasking Endothelial Protein NRG-1 as a Compensatory Factor During Chronic Heart Failure. Circ Heart Fail 2019; 12:e006288. [DOI: 10.1161/circheartfailure.119.006288] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heart failure is a complex syndrome whose phenotypic presentation and disease progression depends on a complex network of adaptive and maladaptive responses. One of these responses is the endothelial release of NRG (neuregulin)-1—a paracrine growth factor activating ErbB2 (erythroblastic leukemia viral oncogene homolog B2), ErbB3, and ErbB4 receptor tyrosine kinases on various targets cells. NRG-1 features a multitasking profile tuning regenerative, inflammatory, fibrotic, and metabolic processes. Here, we review the activities of NRG-1 on different cell types and organs and their implication for heart failure progression and its comorbidities. Although, in general, effects of NRG-1 in heart failure are compensatory and beneficial, translation into therapies remains unaccomplished both because of the complexity of the underlying pathways and because of the challenges in the development of therapeutics (proteins, peptides, small molecules, and RNA-based therapies) for tyrosine kinase receptors. Here, we give an overview of the complexity to be faced and how it may be tackled.
Collapse
Affiliation(s)
- Gilles W. De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium (G.W.D.K.)
| | - Eline Feyen
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Lindsey Dugaucquier
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Anastasia Shchendrygina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation (A.S., Y.N.B.)
| | - Yury N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation (A.S., Y.N.B.)
| | - Marijke Brink
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland (M.B.)
| | - Zarha Vermeulen
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Vincent F. M. Segers
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
- Department of Cardiology, University Hospital Antwerp, Edegem, Belgium (V.F.M.S.)
| |
Collapse
|
24
|
Shakeri H, Gevaert AB, Schrijvers DM, De Meyer GRY, De Keulenaer GW, Guns PJDF, Lemmens K, Segers VF. Neuregulin-1 attenuates stress-induced vascular senescence. Cardiovasc Res 2019. [PMID: 29528383 DOI: 10.1093/cvr/cvy059] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aims Cardiovascular ageing is a key determinant of life expectancy. Cellular senescence, a state of irreversible cell cycle arrest, is an important contributor to ageing due to the accumulation of damaged cells. Targeting cellular senescence could prevent age-related cardiovascular diseases. In this study, we investigated the effects of neuregulin-1 (NRG-1), an epidermal growth factor with cardioprotective and anti-atherosclerotic effects, on cellular senescence. Methods and results Senescence was induced in cultured rat aortic endothelial cells (ECs) and aortic smooth muscle cells (SMCs) by 2 h exposure to 30 µM hydrogen peroxide (H2O2). Cellular senescence was confirmed after 72 h using senescence-associated-β-galactosidase staining (SA-β-gal), cell surface area, and western blot analyses of SA pathways (acetyl-p53, p21). Recombinant human NRG-1 (rhNRG-1, 20 ng/mL) significantly reduced H2O2-induced senescence, as shown by a lower number of SA-β-gal positive cells, smaller surface area and lower expression of acetyl-p53. In C57BL/6 male mice rendered diabetic with streptozotocin (STZ), rhNRG-1 attenuated cellular senescence in aortic ECs and SMCs. Next, we created mice with SMC-specific knockdown of the NRG-1 receptor ErbB4. Aortic SMCs isolated from SMC-specific ErbB4 deficient mice (ErbB4f/+ SM22α-Cre+) showed earlier cellular senescence in vitro compared with wild-type (ErbB4+/+ SM22α-Cre+) SMCs. Furthermore, when rendered diabetic with STZ, ErbB4f/+ SM22α-Cre+ male mice showed significantly more vascular senescence than their diabetic wild-type littermates and had increased mortality. Conclusions This study is the first to explore the role of NRG-1 in vascular senescence. Our data demonstrate that NRG-1 markedly inhibits stress-induced premature senescence in vascular cells in vitro and in the aorta of diabetic mice in vivo. Consistently, deficiency in the NRG-1 receptor ErbB4 provokes cellular senescence in vitro as well as in vivo.
Collapse
Affiliation(s)
- Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Andreas B Gevaert
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.,Department of Cardiology.,Laboratory for Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital (UZA), Antwerp, Belgium
| | - Dorien M Schrijvers
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Pieter-Jan D F Guns
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Katrien Lemmens
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Vincent F Segers
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.,Department of Cardiology
| |
Collapse
|
25
|
Growth factor delivery: Defining the next generation platforms for tissue engineering. J Control Release 2019; 306:40-58. [DOI: 10.1016/j.jconrel.2019.05.028] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
|
26
|
van der Kemp J, van der Schouw YT, Asselbergs FW, Onland-Moret NC. Women-specific risk factors for heart failure: A genetic approach. Maturitas 2018; 109:104-111. [DOI: 10.1016/j.maturitas.2017.12.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023]
|