1
|
Yang Q, Zhong QM, Song MQ, Tong LG, Bai CZ. Exosomes derived from Danshen decoction-pretreated bone marrow mesenchymal stem cells alleviate myocardial infarction via anti-apoptosis and up-regulation of autophagy. Heliyon 2024; 10:e38034. [PMID: 39347388 PMCID: PMC11437974 DOI: 10.1016/j.heliyon.2024.e38034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/12/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Cardiomyocyte loss and myocardial fibrosis are major determinants of myocardial infarction (MI) pathological changes. Mesenchymal stem cell (MSC)-derived exosomes (exos) and Danshen decoction (DSY) have been demonstrated to mediate cardiac repair following MI. BM-MSCs exos or BM-MSCsDSY exos were intramuscularly injected into post-MI rats. On the 7th, 14th and 28th days, serum CK, LDH, α-HBDH, ALT, and AST were measured and electrocardiogram changes were monitored to identify cardiac function; Triphenyltetrazolium chloride staining, Hematein&Eosin staining, Masson trichrome staining and Transmission Electron Microscope were adopted to analyze infarct area, cardiac morphology, histopathology, and fibrosis and cardiomyocyte ultrastructure; TUNEL assay, real-time PCR and western blot were performed to detect cardiomyocyte apoptosis and autophagy. As a result, BMMSCsDSY exos are superior to BM-MSCs-exos in improvement of cardiac function, morphology, histopathology and cardiomyocyte ultrastructure, as well as in reduction of infarction area and cardiac fibrosis by inhibiting apoptosis and promoting autophagy of cardiomyocytes.
Collapse
Affiliation(s)
- Qian Yang
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, No. 70, Nanshifang Street, Taiyuan City, Shanxi Province, 030012, China
| | - Qi-Ming Zhong
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, No. 70, Nanshifang Street, Taiyuan City, Shanxi Province, 030012, China
| | - Mei-Qing Song
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, No. 70, Nanshifang Street, Taiyuan City, Shanxi Province, 030012, China
| | - Li-Guo Tong
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, No. 70, Nanshifang Street, Taiyuan City, Shanxi Province, 030012, China
| | - Chong-Zhi Bai
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, No. 70, Nanshifang Street, Taiyuan City, Shanxi Province, 030012, China
| |
Collapse
|
2
|
Bajic Z, Sobot T, Amidzic L, Vojinovic N, Jovicic S, Gajic Bojic M, Djuric DM, Stojiljkovic MP, Bolevich S, Skrbic R. Liraglutide Protects Cardiomyocytes against Isoprenaline-Induced Apoptosis in Experimental Takotsubo Syndrome. Biomedicines 2024; 12:1207. [PMID: 38927414 PMCID: PMC11200478 DOI: 10.3390/biomedicines12061207] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 06/28/2024] Open
Abstract
Takotsubo syndrome (TTS) is a stress-induced cardiomyopathy, characterized by an increased concentration of catecholamines, free radicals, and inflammatory cytokines, endothelial dysfunction, and increased apoptotic activity. High doses of isoprenaline are used in animal models to induce Takotsubo (TT)-like myocardial injury. The aim of the study was to investigate the antiapoptotic effects of liraglutide in experimental TTS and its role in the NF-κB pathway. Wistar rats were pretreated with liraglutide for 10 days, and on days 9 and 10, TT-like myocardial injury was induced with isoprenaline. After the sacrifice on day 11, hearts were isolated for histopathological and immunohistochemical analysis. Liraglutide reduced isoprenaline-induced cardiomyocyte apoptosis by decreasing cleaved caspase-3 (CC3), BCL-2-associated X protein (BAX), and NF-κB and increasing B-cell lymphoma/leukemia-2 (BCL-2). An increase in NF-κB in isoprenaline-treated rats was in positive correlation with proapoptotic markers (BAX and CC3) and in negative correlation with antiapoptotic marker BCL-2. Liraglutide increased BCL-2 and decreased NF-κB, BAX, and CC3, preserving the same correlations of NF-κB to apoptotic markers. It is concluded that liraglutide protects cardiomyocytes against isoprenaline-induced apoptosis in experimental TT-like myocardial injury through downregulation of the NF-κB pathway.
Collapse
Affiliation(s)
- Zorislava Bajic
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina;
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
| | - Tanja Sobot
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina;
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
| | - Ljiljana Amidzic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Biology of Cell and Human Genetics, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Natasa Vojinovic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Biology of Cell and Human Genetics, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Sanja Jovicic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Histology and Embryology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Milica Gajic Bojic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Dragan M. Djuric
- Faculty of Medicine, Institute of Medical Physiology “Richard Burian”, University of Belgrade, 11 000 Belgrade, Serbia;
| | - Milos P. Stojiljkovic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Sergey Bolevich
- Department of Pathologic Physiology, First Moscow State Medical University I.M. Sechenov, 119435 Moscow, Russia;
| | - Ranko Skrbic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
- Department of Pathologic Physiology, First Moscow State Medical University I.M. Sechenov, 119435 Moscow, Russia;
| |
Collapse
|
3
|
Azhar G, Nagano K, Patyal P, Zhang X, Verma A, Wei JY. Deletion of Interleukin-1β Converting Enzyme Alters Mouse Cardiac Structure and Function. BIOLOGY 2024; 13:172. [PMID: 38534442 DOI: 10.3390/biology13030172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024]
Abstract
Interleukin-1β converting enzyme (ICE, caspase-1) is a thiol protease that cleaves the pro-inflammatory cytokine precursors of IL-1β and IL-18 into active forms. Given the association between caspase-1 and cardiovascular pathology, we analyzed the hearts of ICE knockout (ICE KO) mice to test the hypothesis that caspase-1 plays a significant role in cardiac morphology and function. We characterized the histological and functional changes in the hearts of ICE KO mice compared to the Wild type. The cardiomyocytes from the neonatal ICE KO mice showed an impaired response to oxidative stress. Subsequently, the hearts from the ICE KO mice were hypertrophied, with a significant increase in the left ventricular and septal wall thickness and a greater LV mass/body weight ratio. The ICE KO mice hearts exhibited irregular myofibril arrangements and disruption of the cristae in the mitochondrial structure. Proapoptotic proteins that were significantly increased in the hearts of ICE KO versus the Wild type included pErk, pJNK, p53, Fas, Bax, and caspase 3. Further, the antiapoptotic proteins Bag-1 and Bcl-2 are activated in ICE KO hearts. Functionally, there was an increase in the left ventricular epicardial diameter and volume in ICE KO. In conclusion, our findings support the important role of caspase-1 in maintaining cardiac health; specifically, a significant decrease in caspase-1 is detrimental to the cardiovascular system.
Collapse
Affiliation(s)
- Gohar Azhar
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Koichiro Nagano
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Pankaj Patyal
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xiaomin Zhang
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ambika Verma
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
4
|
Tian L, Jarrah M, Herz H, Chu Y, Xu Y, Tang Y, Yuan J, Mokadem M. Toll-like Receptor 4 Differentially Modulates Cardiac Function in Response to Chronic Exposure to High-Fat Diet and Pressure Overload. Nutrients 2023; 15:5139. [PMID: 38140398 PMCID: PMC10747341 DOI: 10.3390/nu15245139] [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: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND/AIM The impact of myocardial stressors such as high-fat diet (HFD) and pressure overload has been extensively studied. Toll-like receptor 4 (TLR4) deficiency has been suggested to have a protective role in response to these stressors, although some conflicting data exist. Furthermore, there is limited information about the role of TLR4 on cardiac remodeling in response to long-term exposure to stressors. This study aims to investigate the effects of TLR4 deficiency on cardiac histology and physiology in response to chronic stressors. METHODS TLR4-deficient (TLR4-/-) and wild-type (WT) mice were subjected to either HFD or a normal diet (ND) for 28 weeks. Another group underwent abdominal aortic constriction (AAC) or a sham procedure and was monitored for 12 weeks. Inflammatory markers, histology, and echocardiography were used to assess the effects of these interventions. RESULTS TLR4-/- mice exhibited reduced cardiac hypertrophy and fibrosis after long-term HFD exposure compared to ND without affecting cardiac function. On the other hand, TLR4 deficiency worsened cardiac function in response to AAC, leading to decreased ejection fraction (EF%) and increased end-systolic volume (ESV). CONCLUSIONS TLR4 deficiency provided protection against HFD-induced myocardial inflammation but impaired hemodynamic cardiac function under pressure overload conditions. These findings highlight the crucial role of TLR4 and its downstream signaling pathway in maintaining cardiac output during physiologic cardiac hypertrophy in response to pressure overload.
Collapse
Affiliation(s)
- Liping Tian
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Mohammad Jarrah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Hussein Herz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Yi Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
| | - Ying Xu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining 272067, China
| | - Mohamad Mokadem
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA (Y.C.)
- Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA 52242, USA
- Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA 52242, USA
- Iowa City Veterans Affairs Health Care System, Iowa City, IA 52242, USA
| |
Collapse
|
5
|
Li S, Tao G. Perish in the Attempt: Regulated Cell Death in Regenerative and Nonregenerative Tissue. Antioxid Redox Signal 2023; 39:1053-1069. [PMID: 37218435 PMCID: PMC10715443 DOI: 10.1089/ars.2022.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
Significance: A cell plays its roles throughout its life span, even during its demise. Regulated cell death (RCD) is one of the key topics in modern biomedical studies. It is considered the main approach for removing stressed and/or damaged cells. Research during the past two decades revealed more roles of RCD, such as coordinating tissue development and driving compensatory proliferation during tissue repair. Recent Advances: Compensatory proliferation, initially identified in primitive organisms during the regeneration of lost tissue, is an evolutionarily conserved process that also functions in mammals. Among various types of RCD, apoptosis is considered the top candidate to induce compensatory proliferation in damaged tissue. Critical Issues: The roles of apoptosis in the recovery of nonregenerative tissue are still vague. The roles of other types of RCD, such as necroptosis and ferroptosis, have not been well characterized in the context of tissue regeneration. Future Directions: In this review article, we attempt to summarize the recent insights on the role of RCD in tissue repair. We focus on apoptosis, with expansion to ferroptosis and necroptosis, in primitive organisms with significant regenerative capacity as well as common mammalian research models. After gathering hints from regenerative tissue, in the second half of the review, we take a notoriously nonregenerative tissue, the myocardium, as an example to discuss the role of RCD in terminally differentiated quiescent cells. Antioxid. Redox Signal. 39, 1053-1069.
Collapse
Affiliation(s)
- Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
6
|
Yuan Hsieh DJ, Islam MN, Kuo WW, Shibu MA, Lai CH, Lin PY, Lin SZ, Chen MYC, Huang CY. A combination of isoliquiritigenin with Artemisia argyi and Ohwia caudata water extracts attenuates oxidative stress, inflammation, and apoptosis by modulating Nrf2/Ho-1 signaling pathways in SD rats with doxorubicin-induced acute cardiotoxicity. ENVIRONMENTAL TOXICOLOGY 2023; 38:3026-3042. [PMID: 37661764 DOI: 10.1002/tox.23936] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/30/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023]
Abstract
Ohwia caudata (Thunb.) H. Ohashi (Leguminosae) also called as "Evergreen shrub" and Artemisia argyi H.Lév. and Vaniot (Compositae) also named as "Chinese mugwort" those two-leaf extracts frequently used as herbal medicine, especially in south east Asia and eastern Asia. Anthracyclines such as doxorubicin (DOX) are commonly used as effective chemotherapeutic drugs in anticancer therapy around the world. However, chemotherapy-induced cardiotoxicity, dilated cardiomyopathy, and congestive heart failure are seen in patients who receive DOX therapy, with the mechanisms underlying DOX-induced cardiac toxicity remaining unclear. Mitochondrial dysfunction, oxidative stress, inflammatory response, and cardiomyocytes have been shown to play crucial roles in DOX-induced cardiotoxicity. Isoliquiritigenin (ISL, 10 mg/kg) is a bioactive flavonoid compound with protective effects against inflammation, neurodegeneration, cancer, and diabetes. Here, in this study, our aim is to find out the Artemisia argyi (AA) and Ohwia caudata (OC) leaf extract combination with Isoliquiritigenin in potentiating and complementing effect against chemo drug side effect to ameliorate cardiac damage and improve the cardiac function. In this study, we showed that a combination of low (AA 300 mg/kg; OC 100 mg/kg) and high-dose(AA 600 mg/kg; OC 300 mg/kg) AA and OC water extract with ISL activated the cell survival-related AKT/PI3K signaling pathway in DOX-treated cardiac tissue leading to the upregulation of the antioxidant markers SOD, HO-1, and Keap-1 and regulated mitochondrial dysfunction through the Nrf2 signaling pathway. Moreover, the water extract of AA and OC with ISL inhibited the inflammatory response genes IL-6 and IL-1β, possibly through the NFκB/AKT/PI3K/p38α/NRLP3 signaling pathways. The water extract of AA and OC with ISL could be a potential herbal drug treatment for cardiac hypertrophy, inflammatory disease, and apoptosis, which can lead to sudden heart failure.
Collapse
Affiliation(s)
- Dennis Jine Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Md Nazmul Islam
- Cardiovascular and Mitochondria Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | | | - Chin-Hu Lai
- Division of Cardiovascular Surgery, Department of Surgery, Taichung Armed Force General Hospital, Taichung City, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| | - Pi-Yu Lin
- Buddhist Compassion Relief Tzu Chi Foundation, Hualien, Taiwan
| | - Shinn-Zong Lin
- Buddhist Compassion Relief Tzu Chi Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Michael Yu-Chih Chen
- Department of Cardiology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondria Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung City, Taiwan
| |
Collapse
|
7
|
Brunette S, Sharma A, Bell R, Puente L, Megeney LA. Caspase 3 exhibits a yeast metacaspase proteostasis function that protects mitochondria from toxic TDP43 aggregates. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:157-169. [PMID: 37545643 PMCID: PMC10399456 DOI: 10.15698/mic2023.08.801] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 08/08/2023]
Abstract
Caspase 3 activation is a hallmark of cell death and there is a strong correlation between elevated protease activity and evolving pathology in neurodegenerative disease, such as amyotrophic lateral sclerosis (ALS). At the cellular level, ALS is characterized by protein aggregates and inclusions, comprising the RNA binding protein TDP-43, which are hypothesized to trigger pathogenic activation of caspase 3. However, a growing body of evidence indicates this protease is essential for ensuring cell viability during growth, differentiation and adaptation to stress. Here, we explored whether caspase 3 acts to disperse toxic protein aggregates, a proteostasis activity first ascribed to the distantly related yeast metacaspase ScMCA1. We demonstrate that human caspase 3 can functionally substitute for the ScMCA1 and limit protein aggregation in yeast, including TDP-43 inclusions. Proteomic analysis revealed that disrupting caspase 3 in the same yeast substitution model resulted in detrimental TDP-43/mitochondrial protein associations. Similarly, suppression of caspase 3, in either murine or human skeletal muscle cells, led to accumulation of TDP-43 aggregates and impaired mitochondrial function. These results suggest that caspase 3 is not inherently pathogenic, but may act as a compensatory proteostasis factor, to limit TDP-43 protein inclusions and protect organelle function in aggregation related degenerative disease.
Collapse
Affiliation(s)
- Steve Brunette
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Anupam Sharma
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Ryan Bell
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Lawrence Puente
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Lynn A Megeney
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
8
|
Papini G, Furini G, Matteucci M, Biemmi V, Casieri V, Di Lascio N, Milano G, Chincoli LR, Faita F, Barile L, Lionetti V. Cardiomyocyte-targeting exosomes from sulforaphane-treated fibroblasts affords cardioprotection in infarcted rats. J Transl Med 2023; 21:313. [PMID: 37161563 PMCID: PMC10169450 DOI: 10.1186/s12967-023-04155-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Exosomes (EXOs), tiny extracellular vesicles that facilitate cell-cell communication, are being explored as a heart failure treatment, although the features of the cell source restrict their efficacy. Fibroblasts the most prevalent non-myocyte heart cells, release poor cardioprotective EXOs. A noninvasive method for manufacturing fibroblast-derived exosomes (F-EXOs) that target cardiomyocytes and slow cardiac remodeling is expected. As a cardioprotective isothiocyanate, sulforaphane (SFN)-induced F-EXOs (SFN-F-EXOs) should recapitulate its anti-remodeling properties. METHODS Exosomes from low-dose SFN (3 μM/7 days)-treated NIH/3T3 murine cells were examined for number, size, and protein composition. Fluorescence microscopy, RT-qPCR, and western blot assessed cell size, oxidative stress, AcH4 levels, hypertrophic gene expression, and caspase-3 activation in angiotensin II (AngII)-stressed HL-1 murine cardiomyocytes 12 h-treated with various EXOs. The uptake of fluorescently-labeled EXOs was also measured in cardiomyocytes. The cardiac function of infarcted male Wistar rats intramyocardially injected with different EXOs (1·1012) was examined by echocardiography. Left ventricular infarct size, hypertrophy, and capillary density were measured. RESULTS Sustained treatment of NIH/3T3 with non-toxic SFN concentration significantly enhances the release of CD81 + EXOs rich in TSG101 (Tumor susceptibility gene 101) and Hsp70 (Heat Shock Protein 70), and containing maspin, an endogenous histone deacetylase 1 inhibitor. SFN-F-EXOs counteract angiotensin II (AngII)-induced hypertrophy and apoptosis in murine HL-1 cardiomyocytes enhancing SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a) levels more effectively than F-EXOs. In stressed cardiomyocytes, SFN-F-EXOs boost AcH4 levels by 30% (p < 0.05) and significantly reduce oxidative stress more than F-EXOs. Fluorescence microscopy showed that mouse cardiomyocytes take in SFN-F-EXOs ~ threefold more than F-EXOs. Compared to vehicle-injected infarcted hearts, SFN-F-EXOs reduce hypertrophy, scar size, and improve contractility. CONCLUSIONS Long-term low-dose SFN treatment of fibroblasts enhances the release of anti-remodeling cardiomyocyte-targeted F-EXOs, which effectively prevent the onset of HF. The proposed method opens a new avenue for large-scale production of cardioprotective exosomes for clinical application using allogeneic fibroblasts.
Collapse
Affiliation(s)
- Gaia Papini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Giulia Furini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Anesthesiology and Intensive Care Medicine, UOSVD, Fondazione Toscana G. Monasterio, Pisa, Italy
| | - Marco Matteucci
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Vanessa Biemmi
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Svizzera Italiana, 6900, Lugano, Switzerland
| | - Valentina Casieri
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Nicole Di Lascio
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Giuseppina Milano
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lucia Rosa Chincoli
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Lucio Barile
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Svizzera Italiana, 6900, Lugano, Switzerland
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy.
- Anesthesiology and Intensive Care Medicine, UOSVD, Fondazione Toscana G. Monasterio, Pisa, Italy.
| |
Collapse
|
9
|
Guo Z, Liu FY, Yang D, Wang MY, Li CF, Tang N, Ma SQ, An P, Yang Z, Tang QZ. Salidroside ameliorates pathological cardiac hypertrophy via TLR4-TAK1-dependent signaling. Phytother Res 2023; 37:1839-1849. [PMID: 36512326 DOI: 10.1002/ptr.7701] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/09/2022] [Accepted: 11/19/2022] [Indexed: 12/14/2022]
Abstract
Salidroside, a prominent active ingredient in traditional Chinese medicines, is garnering increased attention because of its unique pharmacological effects against ischemic heart disease via MAPK signaling, which plays a critical role in regulating the evolution of ventricular hypertrophy. However, the function of Salidroside on myocardial hypertrophy has not yet been elucidated. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with Salidroside (100 mg kg-1 day-1 ) by oral gavage for 3 weeks starting 1 week after surgery. Four weeks after TAC surgery, the mice were subjected to echocardiography and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes were used to validate the protective effects of Salidroside in response to Angiotensin II (Ang II, 1 μM) stimulation. Here, we proved that Salidroside dramatically inhibited hypertrophic reactions generated by pressure overload and isoproterenol (ISO) injection. Salidroside prevented the activation of the TAK1-JNK/p38 axis. Salidroside pretreatment of TAK1-inhibited cardiomyocytes shows no additional attenuation of Ang II-induced cardiomyocytes hypertrophy and signaling pathway activation. The overexpression of constitutively active TAK1 removed the protective effects of Salidroside on myocardial hypertrophy. TAC-induced increase of TLR4 protein expression was reduced considerably in the Salidroside treated mice. Transient transfection of small interfering RNA targeting TLR4 (siTLR4) in cardiomyocytes did not further decrease the activation of the TAK1/JNK-p38 axis. In conclusion, Salidroside functioned as a TLR4 inhibitor and displayed anti-hypertrophic action via the TAK1/JNK-p38 pathway.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Chen-Fei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
10
|
Gaytan SL, Lawan A, Chang J, Nurunnabi M, Bajpeyi S, Boyle JB, Han SM, Min K. The beneficial role of exercise in preventing doxorubicin-induced cardiotoxicity. Front Physiol 2023; 14:1133423. [PMID: 36969584 PMCID: PMC10033603 DOI: 10.3389/fphys.2023.1133423] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Doxorubicin is a highly effective chemotherapeutic agent widely used to treat a variety of cancers. However, the clinical application of doxorubicin is limited due to its adverse effects on several tissues. One of the most serious side effects of doxorubicin is cardiotoxicity, which results in life-threatening heart damage, leading to reduced cancer treatment success and survival rate. Doxorubicin-induced cardiotoxicity results from cellular toxicity, including increased oxidative stress, apoptosis, and activated proteolytic systems. Exercise training has emerged as a non-pharmacological intervention to prevent cardiotoxicity during and after chemotherapy. Exercise training stimulates numerous physiological adaptations in the heart that promote cardioprotective effects against doxorubicin-induced cardiotoxicity. Understanding the mechanisms responsible for exercise-induced cardioprotection is important to develop therapeutic approaches for cancer patients and survivors. In this report, we review the cardiotoxic effects of doxorubicin and discuss the current understanding of exercise-induced cardioprotection in hearts from doxorubicin-treated animals.
Collapse
Affiliation(s)
- Samantha L. Gaytan
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Ahmed Lawan
- Department of Biological Sciences, College of Science, University of Alabama in Huntsville, Huntsville, AL, United States
| | - Jongwha Chang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, United States
| | - Sudip Bajpeyi
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Jason B. Boyle
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Sung Min Han
- Department of Physiology and Aging, College of Medicine, Institute on Aging, University of Florida, Gainesville, FL, United States
- *Correspondence: Kisuk Min, ; Sung Min Han,
| | - Kisuk Min
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
- *Correspondence: Kisuk Min, ; Sung Min Han,
| |
Collapse
|
11
|
Danciu OC, Holdhoff M, Peterson RA, Fischer JH, Liu LC, Wang H, Venepalli NK, Chowdhery R, Nicholas MK, Russell MJ, Fan TM, Hergenrother PJ, Tarasow TM, Dudek AZ. Phase I study of procaspase-activating compound-1 (PAC-1) in the treatment of advanced malignancies. Br J Cancer 2023; 128:783-792. [PMID: 36470974 PMCID: PMC9977881 DOI: 10.1038/s41416-022-02089-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Procaspase-3 (PC-3) is overexpressed in multiple tumour types and procaspase-activating compound 1 (PAC-1) directly activates PC-3 and induces apoptosis in cancer cells. This report describes the first-in-human, phase I study of PAC-1 assessing maximum tolerated dose, safety, and pharmacokinetics. METHODS Modified-Fibonacci dose-escalation 3 + 3 design was used. PAC-1 was administered orally at 7 dose levels (DL) on days 1-21 of a 28-day cycle. Dose-limiting toxicity (DLT) was assessed during the first two cycles of therapy, and pharmacokinetics analysis was conducted on days 1 and 21 of the first cycle. Neurologic and neurocognitive function (NNCF) tests were performed throughout the study. RESULTS Forty-eight patients were enrolled with 33 completing ≥2 cycles of therapy and evaluable for DLT. DL 7 (750 mg/day) was established as the recommended phase 2 dose, with grade 1 and 2 neurological adverse events noted, while NNCF testing showed stable neurologic and cognitive evaluations. PAC-1's t1/2 was 28.5 h after multi-dosing, and systemic drug exposures achieved predicted therapeutic concentrations. PAC-1 clinical activity was observed in patients with neuroendocrine tumour (NET) with 2/5 patients achieving durable partial response. CONCLUSIONS PAC-1 dose at 750 mg/day was recommended for phase 2 studies. Activity of PAC-1 in treatment-refractory NET warrants further investigation. CLINICAL TRIAL REGISTRATION Clinical Trials.gov: NCT02355535.
Collapse
Affiliation(s)
- Oana C Danciu
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Clinical Trials Office, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Matthias Holdhoff
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | - James H Fischer
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Li C Liu
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, IL, USA
| | - Heng Wang
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, IL, USA
| | - Neeta K Venepalli
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rozina Chowdhery
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - M Kelly Nicholas
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Meredith J Russell
- Clinical Trials Office, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Timothy M Fan
- Vanquish Oncology, Inc., Champaign, IL, USA
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, IL, USA
- Cancer Center at Illinois, Urbana-Champaign, IL, USA
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA
| | - Paul J Hergenrother
- Vanquish Oncology, Inc., Champaign, IL, USA
- Cancer Center at Illinois, Urbana-Champaign, IL, USA
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA
- Department of Chemistry, University of Illinois, Urbana-Champaign, IL, USA
| | | | - Arkadiusz Z Dudek
- HealthPartners Institute, Regions Cancer Care Center, St. Paul, MN, USA
- Vanquish Oncology, Inc., Champaign, IL, USA
| |
Collapse
|
12
|
Limyati Y, Sanjaya A, Lucretia T, Gunadi JW, Biben V, Jasaputra DK, Lesmana R. Potential Role of Exercise in Regulating YAP and TAZ During Cardiomyocytes Aging. Curr Cardiol Rev 2022; 18:24-33. [PMID: 35379136 PMCID: PMC9896415 DOI: 10.2174/1573403x18666220404152924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 01/03/2022] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
Adaptation of cardiac muscle to regular exercise results in morphological and structural changes known as physiological cardiac hypertrophy, to which the Hippo signaling pathway might have contributed. Two major terminal effectors in the Hippo signaling pathway are Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ). The latest studies have reported the role of YAP and TAZ in different life stages, such as in fetal, neonatal, and adult hearts. Their regulation might involve several mechanisms and effectors. One of the possible coregulators is exercise. Exercise plays a role in cardiomyocyte hypertrophic changes during different stages of life, including in aged hearts. YAP/TAZ signaling pathway has a role in physiological cardiac hypertrophy induced by exercise and is associated with cardiac remodelling. Thus, it can be believed that exercise has roles in activating the signaling pathway of YAP and TAZ in aged cardiomyocytes. However, the studies regarding the roles of YAP and TAZ during cardiomyocyte aging are limited. The primary purpose of this review is to explore the response of cardiovascular aging to exercise via signaling pathway of YAP and TAZ.
Collapse
Affiliation(s)
- Yenni Limyati
- Address correspondence to this author at the Postgraduate Doctoral Program Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, 40161; Department of Physical Medicine and Rehabilitation, Unggul Karsa Medika Hospital, Bandung, West Java, 40218; Department of Clinical Skills, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia; Tel/Fax: +62222012186, +62222017621;
| | | | | | | | | | | | | |
Collapse
|
13
|
Tonogai EJ, Huang S, Botham RC, Berry MR, Joslyn SK, Daniel GB, Chen Z, Rao J, Zhang X, Basuli F, Rossmeisl JH, Riggins GJ, LeBlanc AK, Fan TM, Hergenrother PJ. Evaluation of a procaspase-3 activator with hydroxyurea or temozolomide against high-grade meningioma in cell culture and canine cancer patients. Neuro Oncol 2021; 23:1723-1735. [PMID: 34216463 PMCID: PMC8485451 DOI: 10.1093/neuonc/noab161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND High-grade meningioma is an aggressive type of brain cancer that is often recalcitrant to surgery and radiotherapy, leading to poor overall survival. Currently, there are no FDA-approved drugs for meningioma, highlighting the need for new therapeutic options, but development is challenging due to the lack of predictive preclinical models. METHODS To leverage the known overexpression of procaspase-3 in meningioma, PAC-1, a blood-brain barrier penetrant procaspase-3 activator, was evaluated for its ability to induce apoptosis in meningioma cells. To enhance the effects of PAC-1, combinations with either hydroxyurea or temozolomide were explored in cell culture. Both combinations were further investigated in small groups of canine meningioma patients and assessed by MRI, and the novel apoptosis tracer, [18F]C-SNAT4, was evaluated in patients treated with PAC-1 + HU. RESULTS In meningioma cell lines in culture, PAC-1 + HU are synergistic while PAC-1 + TMZ show additive-to-synergistic effects. In canine meningioma patients, PAC-1 + HU led to stabilization of disease and no change in apoptosis within the tumor, whereas PAC-1 + TMZ reduced tumor burden in all three canine patients treated. CONCLUSIONS Our results suggest PAC-1 + TMZ as a potentially efficacious combination for the treatment of human meningioma, and also demonstrate the utility of including pet dogs with meningioma as a means to assess anticancer strategies for this common brain tumor.
Collapse
Affiliation(s)
- Emily J Tonogai
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Shan Huang
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Rachel C Botham
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Matthew R Berry
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | | | - Gregory B Daniel
- Radiology, Department of Small Animal Clinical Sciences, Virgina-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Zixin Chen
- Departments of Radiology and Chemistry, Stanford Medicine, Stanford, California, USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry, Stanford Medicine, Stanford, California, USA
| | - Xiang Zhang
- Chemistry and Synthesis Center, NHLBI, NIH, Bethesda, Maryland, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, NHLBI, NIH, Bethesda, Maryland, USA
| | - John H Rossmeisl
- Neurology and Neurosurgery, Department of Small Animal Clinical Sciences, Virgina-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Gregory J Riggins
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Timothy M Fan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
14
|
He Z, Zeng X, Zhou D, Liu P, Han D, Xu L, Bu T, Wang J, Ke M, Pan X, Du Y, Xue H, Lu D, Luo B. LncRNA Chaer Prevents Cardiomyocyte Apoptosis From Acute Myocardial Infarction Through AMPK Activation. Front Pharmacol 2021; 12:649398. [PMID: 34335241 PMCID: PMC8322763 DOI: 10.3389/fphar.2021.649398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
Long non-coding RNA (lncRNA) is widely reported to be involved in cardiac (patho)physiology. Acute myocardial infarction, in which cardiomyocyte apoptosis plays an important role, is a life-threatening disease. Here, we report the lncRNA Chaer that is anti-apoptotic in cardiomyocytes during Acute myocardial infarction. Importantly, lncRNA Chaer is significantly downregulated in both oxygen-glucose deprivation (oxygen-glucose deprivation)-treated cardiomyocytes in vitro and AMI heart. In vitro, overexpression of lncRNA Chaer with adeno virus reduces cardiomyocyte apoptosis induced by OGD-treated while silencing of lncRNA Chaer increases cardiomyocyte apoptosis instead. In vivo, forced expression of lncRNA Chaer with AAV9 attenuates cardiac apoptosis, reduces infarction area and improves mice heart function in AMI. Interestingly, overexpression of lncRNA Chaer promotes the phosphorylation of AMPK, and AMPK inhibitor Compound C reverses the overexpression of lncRNA Chaer effect of reducing cardiomyocyte apoptosis under OGD-treatment. In summary, we identify the novel ability of lncRNA Chaer in regulating cardiomyocyte apoptosis by promoting phosphorylation of AMPK in AMI.
Collapse
Affiliation(s)
- Zhiyu He
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojun Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Deke Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Traditional Chinese Medicine Hospital of Gaozhou, Department of Cardiology, Gaozhou, China
| | - Peiying Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dunzheng Han
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingling Xu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tong Bu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinping Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengmeng Ke
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiudi Pan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yipeng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hao Xue
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongfeng Lu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bihui Luo
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Yoon JJ, Son CO, Kim HY, Han BH, Lee YJ, Lee HS, Kang DG. Betulinic Acid Protects DOX-Triggered Cardiomyocyte Hypertrophy Response through the GATA-4/Calcineurin/NFAT Pathway. Molecules 2020; 26:E53. [PMID: 33374365 PMCID: PMC7795060 DOI: 10.3390/molecules26010053] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac hypertrophy is a major risk factor for heart failure and leads to cardiovascular morbidity and mortality. Doxorubicin (DOX) is regarded as one of the most potent anthracycline antibiotic agents; however, its clinical usage has some limitations because it has serious cardiotoxic side effects such as dilated cardiomyopathy and congestive heart failure. Betulinic acid (BA) is a pentacyclic-cyclic lupane-type triterpene that has been reported to have anti-bacterial, anti-inflammatory, anti-vascular neogenesis, and anti-fibrotic effects. However, there is no study about its direct effect on DOX induced cardiac hypertrophy and apoptosis. The present study aims to investigate the effect of BA on DOX-induced cardiomyocyte hypertrophy and apoptosis in vitro in H9c2 cells. The H9c2 cells were stimulated with DOX (1 µM) in the presence or absence of BA (0.1-1 μM) and incubated for 24 h. The results of the present study indicated that DOX induces the increase cell surface area and the upregulation of hypertrophy markers including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), beta-myosin heavy chain (β-MHC), and Myosin Light Chain-2 (MLC2) in H9c2 cells. However, the pathological hypertrophic responses were downregulated after BA treatment. Moreover, phosphorylation of JNK, ERK, and p38 in DOX treated H9c2 cells was blocked by BA. As a result of measuring the change in ROS generation using DCF-DA, BA significantly inhibited DOX-induced the production of intracellular reactive oxygen species (ROS) when BA was treated at a concentration of over 0.1 µM. DOX-induced activation of GATA-4 and calcineurin/NFAT-3 signaling pathway were remarkably improved by pre-treating of BA to H9c2 cells. In addition, BA treatment significantly reduced DOX-induced cell apoptosis and protein expression levels of Bax and cleaved caspase-3/-9, while the expression of Bcl-2 was increased by BA. Therefore, BA can be a potential treatment for cardiomyocyte hypertrophy and apoptosis that lead to sudden heart failure.
Collapse
Affiliation(s)
- Jung Joo Yoon
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea; (J.J.Y.); (H.Y.K.); (B.H.H.); (Y.J.L.)
- College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea
| | - Chan Ok Son
- Department of Ophthalmology, School of Medicine, Konkuk University, Gwangjin-gu, Seoul 05029, Korea;
| | - Hye Yoom Kim
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea; (J.J.Y.); (H.Y.K.); (B.H.H.); (Y.J.L.)
- College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea
| | - Byung Hyuk Han
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea; (J.J.Y.); (H.Y.K.); (B.H.H.); (Y.J.L.)
- College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea
| | - Yun Jung Lee
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea; (J.J.Y.); (H.Y.K.); (B.H.H.); (Y.J.L.)
- College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea
| | - Ho Sub Lee
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea; (J.J.Y.); (H.Y.K.); (B.H.H.); (Y.J.L.)
- College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea
| | - Dae Gill Kang
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea; (J.J.Y.); (H.Y.K.); (B.H.H.); (Y.J.L.)
- College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk 54538, Korea
| |
Collapse
|
16
|
Vergarajauregui S, Becker R, Steffen U, Sharkova M, Esser T, Petzold J, Billing F, Kapiloff MS, Schett G, Thievessen I, Engel FB. AKAP6 orchestrates the nuclear envelope microtubule-organizing center by linking golgi and nucleus via AKAP9. eLife 2020; 9:61669. [PMID: 33295871 PMCID: PMC7725499 DOI: 10.7554/elife.61669] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/03/2020] [Indexed: 12/31/2022] Open
Abstract
The switch from centrosomal microtubule-organizing centers (MTOCs) to non-centrosomal MTOCs during differentiation is poorly understood. Here, we identify AKAP6 as key component of the nuclear envelope MTOC. In rat cardiomyocytes, AKAP6 anchors centrosomal proteins to the nuclear envelope through its spectrin repeats, acting as an adaptor between nesprin-1α and Pcnt or AKAP9. In addition, AKAP6 and AKAP9 form a protein platform tethering the Golgi to the nucleus. Both Golgi and nuclear envelope exhibit MTOC activity utilizing either AKAP9, or Pcnt-AKAP9, respectively. AKAP6 is also required for formation and activity of the nuclear envelope MTOC in human osteoclasts. Moreover, ectopic expression of AKAP6 in epithelial cells is sufficient to recruit endogenous centrosomal proteins. Finally, AKAP6 is required for cardiomyocyte hypertrophy and osteoclast bone resorption activity. Collectively, we decipher the MTOC at the nuclear envelope as a bi-layered structure generating two pools of microtubules with AKAP6 as a key organizer.
Collapse
Affiliation(s)
- Silvia Vergarajauregui
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert Becker
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ulrike Steffen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maria Sharkova
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tilman Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jana Petzold
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Billing
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael S Kapiloff
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, Palo Alto, United States
| | - George Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ingo Thievessen
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Erlangen, Germany
| |
Collapse
|
17
|
Boudreau MW, Peh J, Hergenrother PJ. Procaspase-3 Overexpression in Cancer: A Paradoxical Observation with Therapeutic Potential. ACS Chem Biol 2019; 14:2335-2348. [PMID: 31260254 PMCID: PMC6858495 DOI: 10.1021/acschembio.9b00338] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many anticancer strategies rely on the promotion of apoptosis in cancer cells as a means to shrink tumors. Crucial for apoptotic function are executioner caspases, most notably caspase-3, that proteolyze a variety of proteins, inducing cell death. Paradoxically, overexpression of procaspase-3 (PC-3), the low-activity zymogen precursor to caspase-3, has been reported in a variety of cancer types. Until recently, this counterintuitive overexpression of a pro-apoptotic protein in cancer has been puzzling. Recent studies suggest subapoptotic caspase-3 activity may promote oncogenic transformation, a possible explanation for the enigmatic overexpression of PC-3. Herein, the overexpression of PC-3 in cancer and its mechanistic basis is reviewed; collectively, the data suggest the potential for exploitation of PC-3 overexpression with PC-3 activators as a targeted anticancer strategy.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| |
Collapse
|
18
|
Sáez ME, González-Pérez A, Hernández-Olasagarre B, Beà A, Moreno-Grau S, de Rojas I, Monté-Rubio G, Orellana A, Valero S, Comella JX, Sanchís D, Ruiz A. Genome Wide Meta-Analysis identifies common genetic signatures shared by heart function and Alzheimer's disease. Sci Rep 2019; 9:16665. [PMID: 31723151 PMCID: PMC6853976 DOI: 10.1038/s41598-019-52724-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 09/30/2019] [Indexed: 01/01/2023] Open
Abstract
Echocardiography has become an indispensable tool for the study of heart performance, improving the monitoring of individuals with cardiac diseases. Diverse genetic factors associated with echocardiographic measures have been previously reported. The impact of several apoptotic genes in heart development identified in experimental models prompted us to assess their potential association with human cardiac function. This study aimed at investigating the possible association of variants of apoptotic genes with echocardiographic traits and to identify new genetic markers associated with cardiac function. Genome wide data from different studies were obtained from public repositories. After quality control and imputation, a meta-analysis of individual association study results was performed. Our results confirmed the role of caspases and other apoptosis related genes with cardiac phenotypes. Moreover, enrichment analysis showed an over-representation of genes, including some apoptotic regulators, associated with Alzheimer's disease. We further explored this unexpected observation which was confirmed by genetic correlation analyses. Our findings show the association of apoptotic gene variants with echocardiographic indicators of heart function and reveal a novel potential genetic link between echocardiographic measures in healthy populations and cognitive decline later on in life. These findings may have important implications for preventative strategies combating Alzheimer's disease.
Collapse
Affiliation(s)
- M E Sáez
- Andalusian Bioinformatics Research Centre (CAEBi), Seville, Spain
| | - A González-Pérez
- Andalusian Bioinformatics Research Centre (CAEBi), Seville, Spain
| | - B Hernández-Olasagarre
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - A Beà
- Universitat de Lleida - IRBLleida, Lleida, Spain
| | - S Moreno-Grau
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain
| | - I de Rojas
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain
| | - G Monté-Rubio
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - A Orellana
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain
| | - S Valero
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain
| | - J X Comella
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain
- Institut de Recerca Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona, Spain
| | - D Sanchís
- Universitat de Lleida - IRBLleida, Lleida, Spain.
| | - A Ruiz
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.
| |
Collapse
|
19
|
Dronc-independent basal executioner caspase activity sustains Drosophila imaginal tissue growth. Proc Natl Acad Sci U S A 2019; 116:20539-20544. [PMID: 31548372 PMCID: PMC6789915 DOI: 10.1073/pnas.1904647116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Caspase is the enzyme involved in cell death, and its activation via the apoptosome is thought to represent irreversible cellular destruction. Furthermore, accumulating evidence suggests increasingly diverse functions of caspase beyond apoptosis. Here, using Drosophila wing as a model, we reveal that the specific executioner caspases, Dcp-1 and Decay, promote, rather than suppress by inducing apoptosis, tissue growth. These executioner caspases act independently of initiator caspase Dronc and apoptosis. We further show that the caspase-mediated cleavage of Acinus is important for sustaining tissue growth. Our research highlights the importance of executioner caspase-mediated basal proteolytic cleavage of substrates during tissue growth, and the findings hint at the original function of caspase—not apoptosis, but basal proteolytic cleavages for cell vigor. Caspase is best known as an enzyme involved in programmed cell death, which is conserved among multicellular organisms. In addition to its role in cell death, caspase is emerging as an indispensable enzyme in a wide range of cellular functions, which have recently been termed caspase-dependent nonlethal cellular processes (CDPs). In this study, we examined the involvement of cell death signaling in tissue-size determination using Drosophila wing as a model. We found that the Drosophila executioner caspases Dcp-1 and Decay, but not Drice, promoted wing growth independently of apoptosis. Most of the reports on CDPs argue the importance of the spatiotemporal regulation of the initiator caspase, Dronc; however, this sublethal caspase function was independent of Dronc, suggesting a more diverse array of CDP regulatory mechanisms. Tagging of TurboID, an improved promiscuous biotin ligase that biotinylates neighboring proteins, to the C terminus of caspases revealed the differences among the neighbors of executioner caspases. Furthermore, we found that the cleavage of Acinus, a substrate of the executioner caspase, was important in promoting wing growth. These results demonstrate the importance of executioner caspase-mediated basal proteolytic cleavage of substrates in sustaining tissue growth. Given the existence of caspase-like DEVDase activity in a unicellular alga, our results likely highlight the original function of caspase—not cell death, but basal proteolytic cleavages for cell vigor.
Collapse
|
20
|
Li S, Fang J, Chen L. Pyrrolidine Dithiocarbamate Attenuates Cardiocyte Apoptosis and Ameliorates Heart Failure Following Coronary Microembolization in Rats. Balkan Med J 2019; 36:245-250. [PMID: 31140237 PMCID: PMC6636647 DOI: 10.4274/balkanmedj.galenos.2019.2019.3.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/28/2019] [Indexed: 12/01/2022] Open
Abstract
Background Nuclear factor-kB is highly activated in cardiovascular disorders. However, few articles have targeted at the role of nuclear factor-kB inhibitor in heart failure. Aims To evaluate the effects of nuclear factor-kB inhibitor pyrrolidine dithiocarbamate on cardiocyte apoptosis and cardiac function in a rat heart failure model. Study Design Animal experiment. Methods A stable and reproducible rat heart failure model (n=64) was prepared by injecting homologous microthrombotic particles into the left ventricle of Sprague–Dawley rats while obstructing the ascending aorta to produce coronary microembolization. Rats with heart failure were randomized into untreated (HFu) and pyrrolidine dithiocarbamate-treated (HFp) groups; the latter received an intraperitoneal injection of pyrrolidine dithiocarbamate (100 mg/kg/day) 1 h prior to surgery as well as on postoperative days 1-7. The sham group comprised 32 Sprague–Dawley rats. Eight rats from each group were sacrificed on days 1, 3, 7, and 14 postoperatively. Masson’s trichrome staining was used to determine the micro-fibrotic area to indicate the severity of myocardial loss. Terminal transferase uridine triphosphate nick end labeling staining was used to detect apoptotic cardiomyocytes. Echocardiography and hemodynamics were performed to evaluate left ventricular function. Results Rats with heart failure exhibited pathological changes evidenced by patchy myocardial fibrosis, remarkably elevated severity of myocardial loss, and persistently reduced left ventricular function. At the end of the study, compared with the HFu group, myocardial infarct size was reduced by 28% (p=0.001), cardiocyte apoptosis was suppressed (7.17%±1.47% vs 2.83%±0.75%, p<0.001), cardiac function parameters such as left ventricular ejection fraction (80%±4% vs 61%±6%), left ventricular + dP/dt max (4828±289 vs 2918±76 mmHg.s−1), left ventricular - dP/dt max (4398±269 vs 2481±365 mmHg.s−1), and left ventricular systolic pressure (126±13 vs 100±10 mmHg) were significantly increased, and left ventricular end-diastolic pressure was reduced (18±2 vs 13±1 mmHg) (p<0.001, for all) in the HFu group. Conclusion Our rat model can adequately mimic heart failure via coronary vessel embolization. Moreover, pyrrolidine dithiocarbamate treatment can reduce cardiocyte apoptosis and improve cardiac function, which may be beneficial for patients with heart failure secondary to myocardial infarction.
Collapse
Affiliation(s)
- Shumei Li
- Department of Cardiology, Union Hospital, Fujian Medical University, Fujian, China
| | - Jun Fang
- Department of Cardiology, Union Hospital, Fujian Medical University, Fujian, China
| | - Lianglong Chen
- Department of Cardiology, Union Hospital, Fujian Medical University, Fujian, China
| |
Collapse
|
21
|
Song HY, Kim HM, Mushtaq S, Kim WS, Kim YJ, Lim ST, Byun EB. Gamma-Irradiated Chrysin Improves Anticancer Activity in HT-29 Colon Cancer Cells Through Mitochondria-Related Pathway. J Med Food 2019; 22:713-721. [PMID: 31158040 DOI: 10.1089/jmf.2018.4320] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Irradiation technology can improve the biological activities of natural molecules through a structural modification. This study was conducted to investigate the enhancement of the anticancer effects of chrysin upon exposure to gamma irradiation. Gamma irradiation induces the production of new radiolytic peaks simultaneously with the decrease of the chrysin peak, which increases the cytotoxicity in HT-29 human colon cancer cells. An isolated chrysin derivative (CM1) exhibited a stronger apoptotic effect in HT-29 cells than intact chrysin. The apoptotic characteristics induced by CM1 in HT-29 cells was mediated through the intrinsic signaling pathway, including the excessive production of included reactive oxygen species, the dissipation of the mitochondrial membrane potential, regulation of the B cell lymphoma-2 family, activation of caspase-9, 3, and cleavage of poly (adenosine diphosphate-ribose) polymerase. Our findings suggest that CM1 can be a potential anticancer candidate for the treatment of colon cancer.
Collapse
Affiliation(s)
- Ha-Yeon Song
- 1 Department of Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea.,2 Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, Korea
| | - Hye-Min Kim
- 3 Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Sajid Mushtaq
- 1 Department of Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea.,4 Department of Radiation Biotechnology and Applied Radioisotope Science, Korea University of Science and Technology, Deajeon, Korea
| | - Woo Sik Kim
- 1 Department of Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
| | - Young Jun Kim
- 3 Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Seung-Taik Lim
- 2 Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, Korea
| | - Eui-Baek Byun
- 1 Department of Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
| |
Collapse
|
22
|
PARP1 interacts with HMGB1 and promotes its nuclear export in pathological myocardial hypertrophy. Acta Pharmacol Sin 2019; 40:589-598. [PMID: 30030529 DOI: 10.1038/s41401-018-0044-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/10/2018] [Indexed: 11/09/2022]
Abstract
High-mobility group box 1 (HMGB1) exhibits various functions according to its subcellular location, which is finely conditioned by diverse post-translational modifications, such as acetylation. The nuclear HMGB1 may prevent from cardiac hypertrophy, whereas its exogenous protein is proven to induce hypertrophic response. This present study sought to investigate the regulatory relationships between poly(ADP-ribose) polymerase 1 (PARP1) and HMGB1 in the process of pathological myocardial hypertrophy. Primary-cultured neonatal rat cardiomyocytes (NRCMs) were respectively incubated with three cardiac hypertrophic stimulants, including angiotensin II (Ang II), phenylephrine (PE), and isoproterenol (ISO), and cell surface area and the mRNA expression of hypertrophic biomarkers were measured. the catalytic activity of PARP1 was remarkably enhanced, meanwhile HMGB1 excluded from the nucleus. PARP1 overexpression by infecting with adenovirus PARP1 (Ad-PARP1) promoted the nuclear export of HMGB1, facilitated its secretion outside the cell, aggravated cardiomyocyte hypertrophy, which could be alleviated by HMGB1 overexpression. PE treatment led to the similar results, while that effect was widely depressed by PARP1 silencing or its specific inhibitor AG14361. Moreover, SD rats were intraperitoneally injected with 3-aminobenzamide (3AB, 20 mg/kg every day, a well-established PARP1 inhibitor) 7 days after abdominal aortic constriction (AAC) surgery for 6 weeks, echocardiography and morphometry of the hearts were measured. Pre-treatment of 3AB relieved AAC-caused the translocation of nuclear HMGB1 protein, cardiac hypertrophy, and heart dysfunction. Our research offers a novel evidence that PARP1 combines with HMGB1 and accelerates its translocation from nucleus to cytoplasm, and the course finally causes cardiac hypertrophy.
Collapse
|
23
|
Rebellato P, Kaczynska D, Kanatani S, Rayyes IA, Zhang S, Villaescusa C, Falk A, Arenas E, Hermanson O, Louhivuori L, Uhlén P. The T-type Ca 2+ Channel Ca v3.2 Regulates Differentiation of Neural Progenitor Cells during Cortical Development via Caspase-3. Neuroscience 2019; 402:78-89. [PMID: 30677486 DOI: 10.1016/j.neuroscience.2019.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/11/2018] [Accepted: 01/12/2019] [Indexed: 01/02/2023]
Abstract
Here we report that the low-voltage-dependent T-type calcium (Ca2+) channel Cav3.2, encoded by the CACNA1H gene, regulates neuronal differentiation during early embryonic brain development through activating caspase-3. At the onset of neuronal differentiation, neural progenitor cells exhibited spontaneous Ca2+ activity. This activity strongly correlated with the upregulation of CACNA1H mRNA. Cells exhibiting robust spontaneous Ca2+ signaling had increased caspase-3 activity unrelated to apoptosis. Inhibition of Cav3.2 by drugs or viral CACNA1H knock down resulted in decreased caspase-3 activity followed by suppressed neurogenesis. In contrast, when CACNA1H was overexpressed, increased neurogenesis was detected. Cortical slices from Cacna1h knockout mice showed decreased spontaneous Ca2+ activity, a significantly lower protein level of cleaved caspase-3, and microanatomical abnormalities in the subventricular/ventricular and cortical plate zones when compared to their respective embryonic controls. In summary, we demonstrate a novel relationship between Cav3.2 and caspase-3 signaling that affects neurogenesis in the developing brain.
Collapse
Affiliation(s)
- Paola Rebellato
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Dagmara Kaczynska
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Shigeaki Kanatani
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Songbai Zhang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Carlos Villaescusa
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ernest Arenas
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Lauri Louhivuori
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
24
|
Putinski C, Abdul-Ghani M, Brunette S, Burgon PG, Megeney LA. Caspase Cleavage of Gelsolin Is an Inductive Cue for Pathologic Cardiac Hypertrophy. J Am Heart Assoc 2018; 7:e010404. [PMID: 30486716 PMCID: PMC6405540 DOI: 10.1161/jaha.118.010404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Cardiac hypertrophy is an adaptive remodeling event that may improve or diminish contractile performance of the heart. Physiologic and pathologic hypertrophy yield distinct outcomes, yet both are dependent on caspase‐directed proteolysis. This suggests that each form of myocardial growth may derive from a specific caspase cleavage event(s). We examined whether caspase 3 cleavage of the actin capping/severing protein gelsolin is essential for the development of pathologic hypertrophy. Methods and Results Caspase targeting of gelsolin was established through protein analysis of hypertrophic cardiomyocytes and mass spectrometry mapping of cleavage sites. Pathologic agonists induced late‐stage caspase‐mediated cleavage of gelsolin. The requirement of caspase‐mediated gelsolin cleavage for hypertrophy induction was evaluated in primary cardiomyocytes by cell size analysis, monitoring of prohypertrophy markers, and measurement of hypertrophy‐related transcription activity. The in vivo impact of caspase‐mediated cleavage was investigated by echo‐guided intramyocardial injection of adenoviral‐expressed gelsolin. Expression of the N‐terminal gelsolin caspase cleavage fragment was necessary and sufficient to cause pathologic remodeling in isolated cardiomyocytes and the intact heart, whereas expression of a noncleavable form prevents cardiac remodeling. Alterations in myocardium structure and function were determined by echocardiography and end‐stage cardiomyocyte cell size analysis. Gelsolin secretion was also monitored for its impact on naïve cells using competitive antibody trapping, demonstrating that hypertrophic agonist stimulation of cardiomyocytes leads to gelsolin secretion, which induces hypertrophy in naïve cells. Conclusions These results suggest that cell autonomous caspase cleavage of gelsolin is essential for pathologic hypertrophy and that cardiomyocyte secretion of gelsolin may accelerate this negative remodeling response.
Collapse
Affiliation(s)
- Charis Putinski
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Mohammad Abdul-Ghani
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Steve Brunette
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada
| | - Patrick G Burgon
- 2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada.,4 University of Ottawa Heart Institute Ottawa Ontario Canada
| | - Lynn A Megeney
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada
| |
Collapse
|
25
|
A P, P SR, M PR, K G R. Apoptosis in angiotensin II-stimulated hypertrophic cardiac cells -modulation by phenolics rich extract of Boerhavia diffusa L. Biomed Pharmacother 2018; 108:1097-1104. [PMID: 30372810 DOI: 10.1016/j.biopha.2018.09.114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 02/04/2023] Open
Abstract
Herein, we investigated the effects of B. diffusa (BDE), a well-known cardiotonic edible medicinal plant against apoptosis in Angiotensin II (Ang II)-stimulated hypertrophic cardiac cells (H9c2). The cells were analyzed for viability, markers of hypertrophy, apoptosis, and the expression of various proteins related to apoptosis. Ang II (100 nM for 48 h)-exposed H9c2 cells treated with BDE (75 μg/ml) showed a significant reduction in apoptosis (58.60%↓) compared to Ang II-alone treated cells. BDE treatment significantly reduced the up-regulation of Bax and cytosolic cytochrome-C caused by Ang II as well as reduced the degree of Ang II- induced down-regulation of Bcl-2. A reduction in caspase-3 activity (33.77%↓) and down-regulation of TNF-α was also observed in BDE treated cells stimulated with Ang II. Furthermore, the up-regulation of phospho-p38 MAPK was attenuated by BDE treatment. Bioactive components in the extract were identified as boeravinone B, quercetin, kaempferol, and caffeic acid as evident from high-performance liquid chromatography (HPLC). Overall, our study shows that B. diffusa is effective in attenuating apoptosis in cardiac cells, which is a major contributor to sudden cardiac death in addition to its nutraceutical properties.
Collapse
Affiliation(s)
- Prathapan A
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India
| | - Salin Raj P
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India
| | - Priya Rani M
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India
| | - Raghu K G
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India.
| |
Collapse
|
26
|
Yan K, Ponnusamy M, Xin Y, Wang Q, Li P, Wang K. The role of K63-linked polyubiquitination in cardiac hypertrophy. J Cell Mol Med 2018; 22:4558-4567. [PMID: 30102008 PMCID: PMC6156430 DOI: 10.1111/jcmm.13669] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/20/2018] [Indexed: 12/26/2022] Open
Abstract
Ubiquitination, also known as ubiquitylation, is a vital post‐translational modification of proteins that play a crucial role in the multiple biological processes including cell growth, proliferation and apoptosis. K63‐linked ubiquitination is one of the vital post‐translational modifications of proteins that are involved in the activation of protein kinases and protein trafficking during cell survival and proliferation. It also contributes to the development of various disorders including cancer, neurodegeneration and cardiac hypertrophy. In this review, we summarize the role of K63‐linked ubiquitination signalling in protein kinase activation and its implications in cardiac hypertrophy. We have also provided our perspectives on therapeutically targeting K63‐linked ubiquitination in downstream effector molecules of growth factor receptors for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Kaowen Yan
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | | | - Ying Xin
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Wang
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
When dying is not the end: Apoptotic caspases as drivers of proliferation. Semin Cell Dev Biol 2017; 82:86-95. [PMID: 29199139 DOI: 10.1016/j.semcdb.2017.11.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022]
Abstract
Caspases are well known for their role as executioners of apoptosis. However, recent studies have revealed that these lethal enzymes also have important mitogenic functions. Caspases can promote proliferation through autonomous regulation of the cell cycle, as well as by induction of secreted signals, which have a profound impact in neighboring tissues. Here, I review the proliferative role of caspases during development and homeostasis, in addition to their key regenerative function during tissue repair upon injury. Furthermore, the emerging properties of apoptotic caspases as drivers of carcinogenesis are discussed, as well as their involvement in other diseases. Finally, I examine further effects of caspases regulating death and survival in a non-autonomous manner.
Collapse
|
28
|
Burgon PG, Megeney LA. Caspase signaling, a conserved inductive cue for metazoan cell differentiation. Semin Cell Dev Biol 2017; 82:96-104. [PMID: 29129746 DOI: 10.1016/j.semcdb.2017.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Caspase signaling pathways were originally discovered as conveyors of programmed cell death, yet a compendium of research over the past two decades have demonstrated that these same conduits have a plethora of physiologic functions. Arguably the most extensive non-death activity that has been attributed to this protease clade is the capacity to induce cell differentiation. Caspase control of differentiation is conserved across diverse metazoan organisms from flies to humans, suggesting an ancient origin for this form of cell fate control. Here we discuss the mechanisms by which caspase enzymes manage differentiation, the targeted substrates that may be common across cell lineages, and the countervailing signals that may be essential for these proteases to 'execute' this non-death cell fate.
Collapse
Affiliation(s)
- Patrick G Burgon
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Lynn A Megeney
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
29
|
MicroRNA-210-mediated proliferation, survival, and angiogenesis promote cardiac repair post myocardial infarction in rodents. J Mol Med (Berl) 2017; 95:1369-1385. [PMID: 28948298 DOI: 10.1007/s00109-017-1591-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/29/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022]
Abstract
An innovative approach for cardiac regeneration following injury is to induce endogenous cardiomyocyte (CM) cell cycle re-entry. In the present study, CMs from adult rat hearts were isolated and transfected with cel-miR-67 (control) and rno-miR-210. A significant increase in CM proliferation and mono-nucleation were observed in miR-210 group, in addition to a reduction in CM size, multi-nucleation, and cell death. When compared to control, β-catenin and Bcl-2 were upregulated while APC (adenomatous polyposis coli), p16, and caspase-3 were downregulated in miR-210 group. In silico analysis predicted cell cycle inhibitor, APC, as a direct target of miR-210 in rodents. Moreover, compared to control, a significant increase in CM survival and proliferation were observed with siRNA-mediated inhibition of APC. Furthermore, miR-210 overexpressing C57BL/6 mice (210-TG) were used for short-term ischemia/reperfusion study, revealing smaller cell size, increased mono-nucleation, decreased multi-nucleation, and increased CM proliferation in 210-TG hearts in contrast to wild-type (NTG). Likewise, myocardial infarction (MI) was created in adult mice, echocardiography was performed, and the hearts were harvested for immunohistochemistry and molecular studies. Compared to NTG, 210-TG hearts showed a significant increase in CM proliferation, reduced apoptosis, upregulated angiogenesis, reduced infarct size, and overall improvement in cardiac function following MI. β-catenin, Bcl-2, and VEGF (vascular endothelial growth factor) were upregulated while APC, p16, and caspase-3 were downregulated in 210-TG hearts. Overall, constitutive overexpression of miR-210 rescues heart function following cardiac injury in adult mice via promoting CM proliferation, cell survival, and angiogenesis. KEY MESSAGES MiRNA-210 transfected adult rat CMs show proliferation and reduced cell death in vitro. Cell cycle inhibitor APC is a target of miR-210. MiR-210 overexpressing (210-TG) mouse hearts show CMs cell cycle re-entry and survival post myocardial injury. 210-TG mice show significant neovascularization and angiogenic potential post myocardial infarction. 210-TG hearts show reduced infarct size following ischemic injury.
Collapse
|
30
|
Abdul-Ghani M, Suen C, Jiang B, Deng Y, Weldrick JJ, Putinski C, Brunette S, Fernando P, Lee TT, Flynn P, Leenen FHH, Burgon PG, Stewart DJ, Megeney LA. Cardiotrophin 1 stimulates beneficial myogenic and vascular remodeling of the heart. Cell Res 2017; 27:1195-1215. [PMID: 28785017 PMCID: PMC5630684 DOI: 10.1038/cr.2017.87] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/06/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022] Open
Abstract
The post-natal heart adapts to stress and overload through hypertrophic growth, a process that may be pathologic or beneficial (physiologic hypertrophy). Physiologic hypertrophy improves cardiac performance in both healthy and diseased individuals, yet the mechanisms that propagate this favorable adaptation remain poorly defined. We identify the cytokine cardiotrophin 1 (CT1) as a factor capable of recapitulating the key features of physiologic growth of the heart including transient and reversible hypertrophy of the myocardium, and stimulation of cardiomyocyte-derived angiogenic signals leading to increased vascularity. The capacity of CT1 to induce physiologic hypertrophy originates from a CK2-mediated restraining of caspase activation, preventing the transition to unrestrained pathologic growth. Exogenous CT1 protein delivery attenuated pathology and restored contractile function in a severe model of right heart failure, suggesting a novel treatment option for this intractable cardiac disease.
Collapse
Affiliation(s)
- Mohammad Abdul-Ghani
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Colin Suen
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Baohua Jiang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Yupu Deng
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Jonathan J Weldrick
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Charis Putinski
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Steve Brunette
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Pasan Fernando
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Biology, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Tom T Lee
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Peter Flynn
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Frans H H Leenen
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Patrick G Burgon
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Duncan J Stewart
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Lynn A Megeney
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
31
|
Joshi AD, Botham RC, Schlein LJ, Roth HS, Mangraviti A, Borodovsky A, Tyler B, Joslyn S, Looper JS, Podell M, Fan TM, Hergenrother PJ, Riggins GJ. Synergistic and targeted therapy with a procaspase-3 activator and temozolomide extends survival in glioma rodent models and is feasible for the treatment of canine malignant glioma patients. Oncotarget 2017; 8:80124-80138. [PMID: 29113289 PMCID: PMC5655184 DOI: 10.18632/oncotarget.19085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/09/2017] [Indexed: 12/17/2022] Open
Abstract
Purpose Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. Experimental Design Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. Results Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. Conclusions Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Avadhut D Joshi
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel C Botham
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Lisa J Schlein
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Howard S Roth
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Antonella Mangraviti
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alexandra Borodovsky
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Jayme S Looper
- Department of Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Michael Podell
- Department of Neurology, MedVet Chicago, Chicago, IL, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Gregory J Riggins
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Kankeu C, Clarke K, Passante E, Huber HJ. Doxorubicin-induced chronic dilated cardiomyopathy-the apoptosis hypothesis revisited. J Mol Med (Berl) 2016; 95:239-248. [PMID: 27933370 DOI: 10.1007/s00109-016-1494-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/17/2016] [Accepted: 11/25/2016] [Indexed: 01/08/2023]
Abstract
The chemotherapeutic agent doxorubicin (DOX) has significantly increased survival rates of pediatric and adult cancer patients. However, 10% of pediatric cancer survivors will 10-20 years later develop severe dilated cardiomyopathy (DCM), whereby the exact molecular mechanisms of disease progression after this long latency time remain puzzling. We here revisit the hypothesis that elevated apoptosis signaling or its increased likelihood after DOX exposure can lead to an impairment of cardiac function and cause a cardiac dilation. Based on recent literature evidence, we first argue why a dilated phenotype can occur when little apoptosis is detected. We then review findings suggesting that mature cardiomyocytes are protected against DOX-induced apoptosis downstream, but not upstream of mitochondrial outer membrane permeabilisation (MOMP). This lack of MOMP induction is proposed to alter the metabolic phenotype, induce hypertrophic remodeling, and lead to functional cardiac impairment even in the absence of cardiomyocyte apoptosis. We discuss findings that DOX exposure can lead to increased sensitivity to further cardiomyocyte apoptosis, which may cause a gradual loss in cardiomyocytes over time and a compensatory hypertrophic remodeling after treatment, potentially explaining the long lag time in disease onset. We finally note similarities between DOX-exposed cardiomyocytes and apoptosis-primed cancer cells and propose computational system biology as a tool to predict patient individual DOX doses. In conclusion, combining recent findings in rodent hearts and cardiomyocytes exposed to DOX with insights from apoptosis signal transduction allowed us to obtain a molecularly deeper insight in this delayed and still enigmatic pathology of DCM.
Collapse
Affiliation(s)
- Cynthia Kankeu
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Kylie Clarke
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Egle Passante
- School of Pharmacy and Biomedical Sciences, Univ. of Central Lancashire, Preston, UK
| | - Heinrich J Huber
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium. .,Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
33
|
Sun B, Gao L, Ahsan A, Chu P, Song Y, Li H, Zhang Z, Lin Y, Peng J, Song Z, Wang S, Tang Z. Anticancer effect of SZC015 on lung cancer cells through ROS-dependent apoptosis and autophagy induction mechanisms in vitro. Int Immunopharmacol 2016; 40:400-409. [PMID: 27697723 DOI: 10.1016/j.intimp.2016.09.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/31/2016] [Accepted: 09/24/2016] [Indexed: 12/16/2022]
Abstract
Oleanolic acid (OA) and its several derivatives possess various pharmacological activities, such as antitumor and anti-inflammation. In present study, anticancer effect of SZC015, an OA derivative, and its underlying mechanisms were investigated. We demonstrated that cell viability was significantly decreased in SZC015-treated lung cancer cells, but has less cytotoxicity in human bronchial epithelial cell line. Further investigation verified that apoptosis and autophagy induction and G0/G1 phase arrest were observed in SZC015-treated H322 cells. Mechanically, the level of Akt, p-Akt, p-IκBα, and total p65, the p-p65 in the cytoplasm and nucleus were suppressed by SZC015 in H322 cells, respectively. Inhibition of p65 nuclear translocation was also confirmed by immunofluorescence staining. In addition, co-treatment with chloroquine, an autophagy inhibitor, significantly inhibited SZC015-induced autophagy and enhanced SZC015-induced apoptotic cell death. Intracellular ROS was increased in a concentration-dependent manner, which could be prevented by N-Acetyl l-Cysteine, an ROS scavenger. Moreover, the level of Akt and procaspase-3 were increased, while the ratio of LC3 II/I was decreased. Taken together, our study demonstrates that the inhibitory effect of SZC015 against H322 cells is mediated by excessive ROS generation that could suppress Akt/NF-κB signaling pathway, which thereby leads to apoptotic and autophagic cell death.
Collapse
Affiliation(s)
- Bin Sun
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Lei Gao
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Anil Ahsan
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Peng Chu
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Yanlin Song
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Hailong Li
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Zonghui Zhang
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Yuan Lin
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Jinyong Peng
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Zhicheng Song
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Shisheng Wang
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China.
| | - Zeyao Tang
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China.
| |
Collapse
|
34
|
Botham R, Roth HS, Book AP, Roady PJ, Fan TM, Hergenrother PJ. Small-Molecule Procaspase-3 Activation Sensitizes Cancer to Treatment with Diverse Chemotherapeutics. ACS CENTRAL SCIENCE 2016; 2:545-59. [PMID: 27610416 PMCID: PMC4999974 DOI: 10.1021/acscentsci.6b00165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Indexed: 05/08/2023]
Abstract
Conventional chemotherapeutics remain essential treatments for most cancers, but their combination with other anticancer drugs (including targeted therapeutics) is often complicated by unpredictable synergies and multiplicative toxicities. As cytotoxic anticancer chemotherapeutics generally function through induction of apoptosis, we hypothesized that a molecularly targeted small molecule capable of facilitating a central and defining step in the apoptotic cascade, the activation of procaspase-3 to caspase-3, would broadly and predictably enhance activity of cytotoxic drugs. Here we show that procaspase-activating compound 1 (PAC-1) enhances cancer cell death induced by 15 different FDA-approved chemotherapeutics, across many cancer types and chemotherapeutic targets. In particular, the promising combination of PAC-1 and doxorubicin induces a synergistic reduction in tumor burden and enhances survival in murine tumor models of osteosarcoma and lymphoma. This PAC-1/doxorubicin combination was evaluated in 10 pet dogs with naturally occurring metastatic osteosarcoma or lymphoma, eliciting a biologic response in 3 of 6 osteosarcoma patients and 4 of 4 lymphoma patients. Importantly, in both mice and dogs, coadministration of PAC-1 with doxorubicin resulted in no additional toxicity. On the basis of the mode of action of PAC-1 and the high expression of procaspase-3 in many cancers, these results suggest the combination of PAC-1 with cytotoxic anticancer drugs as a potent and general strategy to enhance therapeutic response.
Collapse
Affiliation(s)
- Rachel
C. Botham
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Howard S. Roth
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Alison P. Book
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Patrick J. Roady
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department of Chemistry, Institute for Genomic Biology, Department of Veterinary
Clinical Medicine, and Veterinary Diagnostic
Laboratory, University of Illinois Urbana−Champaign, Urbana, Illinois 61801, United States
- E-mail:
| |
Collapse
|
35
|
Roh J, Rhee J, Chaudhari V, Rosenzweig A. The Role of Exercise in Cardiac Aging: From Physiology to Molecular Mechanisms. Circ Res 2016; 118:279-95. [PMID: 26838314 DOI: 10.1161/circresaha.115.305250] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aging induces structural and functional changes in the heart that are associated with increased risk of cardiovascular disease and impaired functional capacity in the elderly. Exercise is a diagnostic and therapeutic tool, with the potential to provide insights into clinical diagnosis and prognosis, as well as the molecular mechanisms by which aging influences cardiac physiology and function. In this review, we first provide an overview of how aging impacts the cardiac response to exercise, and the implications this has for functional capacity in older adults. We then review the underlying molecular mechanisms by which cardiac aging contributes to exercise intolerance, and conversely how exercise training can potentially modulate aging phenotypes in the heart. Finally, we highlight the potential use of these exercise models to complement models of disease in efforts to uncover new therapeutic targets to prevent or treat heart disease in the aging population.
Collapse
Affiliation(s)
- Jason Roh
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston
| | - James Rhee
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Vinita Chaudhari
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Anthony Rosenzweig
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston.
| |
Collapse
|
36
|
Peh J, Fan TM, Wycislo KL, Roth HS, Hergenrother PJ. The Combination of Vemurafenib and Procaspase-3 Activation Is Synergistic in Mutant BRAF Melanomas. Mol Cancer Ther 2016; 15:1859-69. [PMID: 27297867 DOI: 10.1158/1535-7163.mct-16-0025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/31/2016] [Indexed: 12/14/2022]
Abstract
The development of vemurafenib resistance limits the long-term efficacy of this drug for treatment of metastatic melanomas with the (V600E)BRAF mutation. Inhibition of downstream MAPK signaling with vemurafenib induces apoptotic cell death mediated by caspase-3, suggesting that addition of a procaspase-3 activator could enhance anticancer effects. Here, we show that the combination of PAC-1, a procaspase-activating compound, and vemurafenib is highly synergistic in enhancing caspase-3 activity and apoptotic cell death in melanoma cell lines harboring the (V600E)BRAF mutation. In vivo, the combination displays a favorable safety profile in mice and exerts significant antitumor effects. We further demonstrate that addition of PAC-1 to the clinically useful combination of vemurafenib and a MEK inhibitor, trametinib, starkly enhances the caspase-3 activity and proapoptotic effect of the combination. Moreover, addition of low concentration PAC-1 also delays the regrowth of cells following treatment with vemurafenib. Finally, PAC-1 remains potent against vemurafenib-resistant A375VR cells in cell culture and synergizes with vemurafenib to exert antitumor effects on A375VR cell growth in vivo Collectively, our data suggest that inhibition of MAPK signaling combined with concurrent procaspase-3 activation is an effective strategy to enhance the antitumor activity of vemurafenib and mitigate the development of resistance. Mol Cancer Ther; 15(8); 1859-69. ©2016 AACR.
Collapse
Affiliation(s)
- Jessie Peh
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois. Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Timothy M Fan
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois. Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Kathryn L Wycislo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Howard S Roth
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois. Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois. Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
37
|
Ji YX, Zhang P, Zhang XJ, Zhao YC, Deng KQ, Jiang X, Wang PX, Huang Z, Li H. The ubiquitin E3 ligase TRAF6 exacerbates pathological cardiac hypertrophy via TAK1-dependent signalling. Nat Commun 2016; 7:11267. [PMID: 27249171 PMCID: PMC4895385 DOI: 10.1038/ncomms11267] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/07/2016] [Indexed: 12/17/2022] Open
Abstract
Tumour necrosis factor receptor-associated factor 6 (TRAF6) is a ubiquitin E3 ligase that regulates important biological processes. However, the role of TRAF6 in cardiac hypertrophy remains unknown. Here, we show that TRAF6 levels are increased in human and murine hypertrophied hearts, which is regulated by reactive oxygen species (ROS) production. Cardiac-specific Traf6 overexpression exacerbates cardiac hypertrophy in response to pressure overload or angiotensin II (Ang II) challenge, whereas Traf6 deficiency causes an alleviated hypertrophic phenotype in mice. Mechanistically, we show that ROS, generated during hypertrophic progression, triggers TRAF6 auto-ubiquitination that facilitates recruitment of TAB2 and its binding to transforming growth factor beta-activated kinase 1 (TAK1), which, in turn, enables the direct TRAF6-TAK1 interaction and promotes TAK1 ubiquitination. The binding of TRAF6 to TAK1 and the induction of TAK1 ubiquitination and activation are indispensable for TRAF6-regulated cardiac remodelling. Taken together, we define TRAF6 as an essential molecular switch leading to cardiac hypertrophy in a TAK1-dependent manner.
Collapse
Affiliation(s)
- Yan-Xiao Ji
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Yi-Chao Zhao
- Department of Cardiology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Ke-Qiong Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xi Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Zan Huang
- College of Life Science, Wuhan University, Wuhan 430072, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| |
Collapse
|
38
|
QiNan W, XiaGuang G, XiaoTian L, WuQuan D, Ling Z, Bing C. Par-4/NF-κB Mediates the Apoptosis of Islet β Cells Induced by Glucolipotoxicity. J Diabetes Res 2016; 2016:4692478. [PMID: 27340675 PMCID: PMC4906207 DOI: 10.1155/2016/4692478] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023] Open
Abstract
Apoptosis of islet β cells is a primary pathogenic feature of type 2 diabetes, and ER stress and mitochondrial dysfunction play important roles in this process. Previous research has shown that prostate apoptosis response-4 (Par-4)/NF-κB induces cancer cell apoptosis through endoplasmic reticulum (ER) stress and mitochondrial dysfunction. However, the mechanism by which Par-4/NF-κB induces islet β cell apoptosis remains unknown. We used a high glucose/palmitate intervention to mimic type 2 diabetes in vitro. We demonstrated that the high glucose/palmitate intervention induced the expression and secretion of Par-4. It also causes increased expression and activation of NF-κB, which induced NIT-1 cell apoptosis and dysfunction. Overexpression of Par-4 potentiates these effects, whereas downregulation of Par-4 attenuates them. Inhibition of NF-κB inhibited the Par-4-induced apoptosis. Furthermore, these effects occurred through the ER stress cell membrane and mitochondrial pathway of apoptosis. Our findings reveal a novel role for Par-4/NF-κB in islet β cell apoptosis and type 2 diabetes.
Collapse
Affiliation(s)
- Wu QiNan
- Endocrine Department, The First Affiliated Hospital of the Third Military Medical University, Chong Qing 400038, China
| | - Gan XiaGuang
- Endocrine Department, The First Affiliated Hospital of the Third Military Medical University, Chong Qing 400038, China
| | - Lei XiaoTian
- Endocrine Department, The First Affiliated Hospital of the Third Military Medical University, Chong Qing 400038, China
| | - Deng WuQuan
- Endocrine Department, The First Affiliated Hospital of the Third Military Medical University, Chong Qing 400038, China
| | - Zhang Ling
- Outpatient Department, The First Affiliated Hospital of the Third Military Medical University, Chong Qing 400038, China
| | - Chen Bing
- Endocrine Department, The First Affiliated Hospital of the Third Military Medical University, Chong Qing 400038, China
| |
Collapse
|
39
|
Bell RAV, Al-Khalaf M, Megeney LA. The beneficial role of proteolysis in skeletal muscle growth and stress adaptation. Skelet Muscle 2016; 6:16. [PMID: 27054028 PMCID: PMC4822268 DOI: 10.1186/s13395-016-0086-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/17/2016] [Indexed: 12/17/2022] Open
Abstract
Muscle atrophy derived from excessive proteolysis is a hallmark of numerous disease conditions. Accordingly, the negative consequences of skeletal muscle protein breakdown often overshadow the critical nature of proteolytic systems in maintaining normal cellular function. Here, we discuss the major cellular proteolysis machinery-the ubiquitin/proteosome system, the autophagy/lysosomal system, and caspase-mediated protein cleavage-and the critical role of these protein machines in establishing and preserving muscle health. We examine how ordered degradation modifies (1) the spatiotemporal expression of myogenic regulatory factors during myoblast differentiation, (2) membrane fusion during myotube formation, (3) sarcomere remodeling and muscle growth following physical stress, and (4) energy homeostasis during nutrient deprivation. Finally, we review the origin and etiology of a number of myopathies and how these devastating conditions arise from inborn errors in proteolysis.
Collapse
Affiliation(s)
- Ryan A V Bell
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6 Canada ; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON Canada
| | - Mohammad Al-Khalaf
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6 Canada ; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON Canada
| | - Lynn A Megeney
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6 Canada ; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON Canada ; Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, ON Canada
| |
Collapse
|
40
|
Pérez-Garijo A, Steller H. Spreading the word: non-autonomous effects of apoptosis during development, regeneration and disease. Development 2016; 142:3253-62. [PMID: 26443630 DOI: 10.1242/dev.127878] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apoptosis, in contrast to other forms of cell death such as necrosis, was originally regarded as a 'silent' mechanism of cell elimination designed to degrade the contents of doomed cells. However, during the past decade it has become clear that apoptotic cells can produce diverse signals that have a profound impact on neighboring cells and tissues. For example, apoptotic cells can release factors that influence the proliferation and survival of adjacent tissues. Apoptosis can also affect tissue movement and morphogenesis by modifying tissue tension in surrounding cells. As we review here, these findings reveal unexpected roles for apoptosis in tissue remodeling during development, as well as in regeneration and cancer.
Collapse
Affiliation(s)
- Ainhoa Pérez-Garijo
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Hermann Steller
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
41
|
Roth HS, Hergenrother PJ. Derivatives of Procaspase-Activating Compound 1 (PAC-1) and their Anticancer Activities. Curr Med Chem 2016; 23:201-41. [PMID: 26630918 PMCID: PMC4968085 DOI: 10.2174/0929867323666151127201829] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/04/2015] [Accepted: 11/27/2015] [Indexed: 01/26/2023]
Abstract
PAC-1 induces the activation of procaspase-3 in vitro and in cell culture by chelation of inhibitory labile zinc ions via its ortho-hydroxy-N-acylhydrazone moiety. First reported in 2006, PAC-1 has shown promise in cell culture and animal models of cancer, and a Phase I clinical trial in cancer patients began in March 2015 (NCT02355535). Because of the considerable interest in this compound and a well-defined structure-activity relationship, over 1000 PAC-1 derivatives have been synthesized in an effort to vary pharmacological properties such as potency and pharmacokinetics. This article provides a comprehensive examination of all PAC-1 derivatives reported to date. A survey of PAC-1 derivative libraries is provided, with an indepth discussion of four derivatives on which extensive studies have been performed.
Collapse
Affiliation(s)
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois, 261 Roger Adams Laboratory, Box 36-5, 600 S. Mathews Ave., Urbana, IL, 61801, USA.
| |
Collapse
|
42
|
Abstract
Myocardial infarction is defined as sudden ischemic death of myocardial tissue. In the clinical context, myocardial infarction is usually due to thrombotic occlusion of a coronary vessel caused by rupture of a vulnerable plaque. Ischemia induces profound metabolic and ionic perturbations in the affected myocardium and causes rapid depression of systolic function. Prolonged myocardial ischemia activates a "wavefront" of cardiomyocyte death that extends from the subendocardium to the subepicardium. Mitochondrial alterations are prominently involved in apoptosis and necrosis of cardiomyocytes in the infarcted heart. The adult mammalian heart has negligible regenerative capacity, thus the infarcted myocardium heals through formation of a scar. Infarct healing is dependent on an inflammatory cascade, triggered by alarmins released by dying cells. Clearance of dead cells and matrix debris by infiltrating phagocytes activates anti-inflammatory pathways leading to suppression of cytokine and chemokine signaling. Activation of the renin-angiotensin-aldosterone system and release of transforming growth factor-β induce conversion of fibroblasts into myofibroblasts, promoting deposition of extracellular matrix proteins. Infarct healing is intertwined with geometric remodeling of the chamber, characterized by dilation, hypertrophy of viable segments, and progressive dysfunction. This review manuscript describes the molecular signals and cellular effectors implicated in injury, repair, and remodeling of the infarcted heart, the mechanistic basis of the most common complications associated with myocardial infarction, and the pathophysiologic effects of established treatment strategies. Moreover, we discuss the implications of pathophysiological insights in design and implementation of new promising therapeutic approaches for patients with myocardial infarction.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
43
|
Abstract
Compensatory growth and regeneration of skeletal muscle is dependent on the resident stem cell population, satellite cells (SCs). Self-renewal and maintenance of the SC niche is coordinated by the paired-box transcription factor Pax7, and yet continued expression of this protein inhibits the myoblast differentiation program. As such, the reduction or removal of Pax7 may denote a key prerequisite for SCs to abandon self-renewal and acquire differentiation competence. Here, we identify caspase 3 cleavage inactivation of Pax7 as a crucial step for terminating the self-renewal process. Inhibition of caspase 3 results in elevated Pax7 protein and SC self-renewal, whereas caspase activation leads to Pax7 cleavage and initiation of the myogenic differentiation program. Moreover, in vivo inhibition of caspase 3 activity leads to a profound disruption in skeletal muscle regeneration with an accumulation of SCs within the niche. We have also noted that casein kinase 2 (CK2)-directed phosphorylation of Pax7 attenuates caspase-directed cleavage. Together, these results demonstrate that SC fate is dependent on opposing posttranslational modifications of the Pax7 protein.
Collapse
|
44
|
Banerjee P, Chander V, Bandyopadhyay A. Balancing functions of annexin A6 maintain equilibrium between hypertrophy and apoptosis in cardiomyocytes. Cell Death Dis 2015; 6:e1873. [PMID: 26335715 PMCID: PMC4650436 DOI: 10.1038/cddis.2015.231] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/09/2015] [Accepted: 07/10/2015] [Indexed: 01/11/2023]
Abstract
Pathological cardiac hypertrophy is a major risk factor associated with heart failure, a state concomitant with increased cell death. However, the mechanism governing progression of hypertrophy to apoptosis at the single-cell level remains elusive. Here, we demonstrate annexin A6 (Anxa6), a calcium (Ca(2+))-dependent phospholipid-binding protein critically regulates the transition of chronic hypertrophied cardiomyocytes to apoptosis. Treatment of the H9c2(2-1) cardiomyocytes with hypertrophic agonists upregulates and relocalizes Anxa6 with increased cytosolic punctate appearance. Live cell imaging revealed that chronic exposure to hypertrophic agonists such as phenylephrine (PE) compromises the mitochondrial membrane potential (ΔΨm) and morphological dynamics. Such chronic hypertrophic induction also activated the caspases 9 and 3 and induced cleavage of the poly-(ADP-ribose) polymerase 1 (Parp1), which are the typical downstream events in the mitochondrial pathways of apoptosis. An increased rate of apoptosis was evident in the hypertrophied cardiomyocytes after 48-72 h of treatment with the hypertrophic agonists. Anxa6 was progressively associated with the mitochondrial fraction under chronic hypertrophic stimulation, and Anxa6 knockdown severely abrogated mitochondrial network and dynamics. Ectopically expressed Anxa6 protected the mitochondrial morphology and dynamics under PE treatment, and also increased the cellular susceptibility to apoptosis. Biochemical analysis showed that Anxa6 interacts with Parp1 and its 89 kDa cleaved product in a Ca(2+)-dependent manner through the N-terminal residues (1-28). Furthermore, expression of Anxa6(S13E), a mutant dominant negative with respect to Parp1 binding, served as an enhancer of mitochondrial dynamics, even under chronic PE treatment. Chemical inhibition of Parp1 activity released the cellular vulnerability to apoptosis in Anxa6-expressing stable cell lines, thereby shifting the equilibrium away from cell death. Taken together, the present study depicts a dual regulatory function of Anxa6 that is crucial for balancing hypertrophy with apoptosis in cardiomyocytes.
Collapse
Affiliation(s)
- P Banerjee
- Cell Biology and Physiology Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Kolkata 700 032, West Bengal, India
| | - V Chander
- Cell Biology and Physiology Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Kolkata 700 032, West Bengal, India
| | - A Bandyopadhyay
- Cell Biology and Physiology Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Kolkata 700 032, West Bengal, India
| |
Collapse
|
45
|
Roth HS, Botham RC, Schmid SC, Fan TM, Dirikolu L, Hergenrother PJ. Removal of Metabolic Liabilities Enables Development of Derivatives of Procaspase-Activating Compound 1 (PAC-1) with Improved Pharmacokinetics. J Med Chem 2015; 58:4046-65. [PMID: 25856364 DOI: 10.1021/acs.jmedchem.5b00413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Procaspase-activating compound 1 (PAC-1) is an o-hydroxy-N-acylhydrazone that induces apoptosis in cancer cells by chelation of labile inhibitory zinc from procaspase-3. PAC-1 has been assessed in a wide variety of cell culture experiments and in vivo models of cancer, with promising results, and a phase 1 clinical trial in cancer patients has been initiated (NCT02355535). For certain applications, however, the in vivo half-life of PAC-1 could be limiting. Thus, with the goal of developing a compound with enhanced metabolic stability, a series of PAC-1 analogues were designed containing modifications that systematically block sites of metabolic vulnerability. Evaluation of the library of compounds identified four potentially superior candidates with comparable anticancer activity in cell culture, enhanced metabolic stability in liver microsomes, and improved tolerability in mice. In head-to-head experiments with PAC-1, pharmacokinetic evaluation in mice demonstrated extended elimination half-lives and greater area under the curve values for each of the four compounds, suggesting them as promising candidates for further development.
Collapse
Affiliation(s)
- Howard S Roth
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Rachel C Botham
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Steven C Schmid
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Timothy M Fan
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Levent Dirikolu
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Paul J Hergenrother
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| |
Collapse
|
46
|
Garcia Barrado MJ, Iglesias Osma MC, Blanco EJ, Carretero Hernández M, Sánchez Robledo V, Catalano Iniesta L, Carrero S, Carretero J. Dopamine modulates insulin release and is involved in the survival of rat pancreatic beta cells. PLoS One 2015; 10:e0123197. [PMID: 25886074 PMCID: PMC4401745 DOI: 10.1371/journal.pone.0123197] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/18/2015] [Indexed: 12/30/2022] Open
Abstract
The local synthesis of dopamine and its effects on insulin release have been described in isolated islets. Thus, it may be accepted that dopamine exerts an auto-paracrine regulation of insulin secretion from pancreatic beta cells. The aim of the present study is to analyze whether dopamine is a regulator of the proliferation and apoptosis of rat pancreatic beta cells after glucose-stimulated insulin secretion. Glucose stimulated pancreatic islets obtained from male Wistar rats were cultured with 1 or 10 μM dopamine from 1 to 12 h. Insulin secretion was analyzed by RIA. The cellular proliferation rate of pancreatic islets and beta cells was studied with immunocytochemical double labelling for both insulin and PCNA (proliferating cell nuclear antigen), and active caspase-3 was detected to evaluate apoptosis. The secretion of insulin from isolated islets was significantly inhibited (p<0.01), by treatment with 1 and 10 μM dopamine, with no differences between either dose as early as 1 h after treatment. The percentage of insulin-positive cells in the islets decreased significantly (p<0.01) after 1 h of treatment up to 12 h. The proliferation rate of insulin-positive cells in the islets decreased significantly (p<0.01) following treatment with dopamine. Apoptosis in pancreatic islets and beta cells was increased by treatment with 1 and 10 μM dopamine along 12 h. In conclusion, these results suggest that dopamine could modulate the proliferation and apoptosis of pancreatic beta cells and that dopamine may be involved in the maintenance of pancreatic islets.
Collapse
Affiliation(s)
- Maria Jose Garcia Barrado
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Maria Carmen Iglesias Osma
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Enrique J. Blanco
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Marta Carretero Hernández
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Virginia Sánchez Robledo
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Leonardo Catalano Iniesta
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Sixto Carrero
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Jose Carretero
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
- * E-mail:
| |
Collapse
|
47
|
Chronic activation of mTOR complex 1 by branched chain amino acids and organ hypertrophy. Amino Acids 2015; 47:1167-82. [PMID: 25721400 DOI: 10.1007/s00726-015-1944-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 02/14/2015] [Indexed: 12/11/2022]
Abstract
The mitochondrial branched chain aminotransferase-deficient mouse model (BCATm KO), which exhibits elevated plasma and tissue branched chain amino acids (BCAAs), was used to study the effect of BCAAs on mammalian target of rapamycin complex 1 (mTORC1) regulation of organ size. BCATm is the first enzyme in the BCAA catabolic pathway. BCATm KO mouse exhibited hypertrophy of heart, kidneys, and spleen. On the other hand, the mass of the gastrocnemius was reduced relative to body mass. Feeding the mice with a diet supplemented with rapamycin prevented the enlargement of the heart and spleen, suggesting that mTORC1 is the mediator of these effects. Consistently, enlargement of these organs was accompanied by the activation of mTORC1 complex as evidenced by enhanced levels of S6 and 4E-BP1 phosphorylation. HSP20, HSP27 and GAPDH were also increased in the heart but not gastrocnemius, consistent with mTORC1 activation. Liver, however, displayed no weight difference between the KO and the wild-type mice despite the highest activation level of mTORC1 complex. These observations suggest that the anabolic effect of mTORC1 activation at the organ level by BCAAs and inhibition by rapamycin are complex phenomenon and tissue-specific. In addition, it suggests that rapamycin can be used to counter hypertrophy of the organs when activation of mTORC1 is the underlying cause.
Collapse
|
48
|
Jain A, Atale N, Kohli S, Bhattacharya S, Sharma M, Rani V. An assessment of norepinephrine mediated hypertrophy to apoptosis transition in cardiac cells: A signal for cell death. Chem Biol Interact 2015; 225:54-62. [DOI: 10.1016/j.cbi.2014.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/18/2014] [Accepted: 11/21/2014] [Indexed: 12/13/2022]
|
49
|
Expression of executioner procaspases and their activation by a procaspase-activating compound in chronic lymphocytic leukemia cells. Blood 2014; 125:1126-36. [PMID: 25538042 DOI: 10.1182/blood-2014-01-546796] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Intrinsic and extrinsic apoptotic pathways converge to activate common downstream executioner caspases (caspase-3, -6, and -7), resulting in cell death. In chronic lymphocytic leukemia (CLL), neoplastic B cells evade apoptosis owing to the overexpression of survival proteins. We hypothesized that direct activation of procaspases could bypass the apoptosis resistance induced by the upstream prosurvival proteins. The procaspase-activating compounds (PAC-1), including B-PAC-1 (L14R8), convert inactive executioner procaspases to their active cleaved forms by chelation of labile zinc ions. Both at transcript and protein levels, primary CLL cells express high levels of latent procaspases (3, -7, and -9). B-PAC-1 treatment induced CLL lymphocyte death which was higher than that in normal peripheral blood mononuclear cells or B cells, and was independent of prognostic markers and microenvironmental factors. Mechanistically, B-PAC-1 treatment activated executioner procaspases and not other Zn-dependent enzymes. Exogenous zinc completely, and pancaspase inhibitors partially, reversed B-PAC-1-induced apoptosis, elucidating the zinc-mediated mechanism of action. The cell demise relied on the presence of caspase-3/7 but not caspase-8 or Bax/Bak proteins. B-PAC-1 in combination with an inhibitor of apoptosis protein antagonist (Smac066) synergistically induced apoptosis in CLL samples. Our investigations demonstrated that direct activation of executioner procaspases via B-PAC-1 treatment bypasses apoptosis resistance and is a novel approach for CLL therapeutics.
Collapse
|
50
|
Differential and conditional activation of PKC-isoforms dictates cardiac adaptation during physiological to pathological hypertrophy. PLoS One 2014; 9:e104711. [PMID: 25116170 PMCID: PMC4130596 DOI: 10.1371/journal.pone.0104711] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/11/2014] [Indexed: 02/02/2023] Open
Abstract
A cardiac hypertrophy is defined as an increase in heart mass which may either be beneficial (physiological hypertrophy) or detrimental (pathological hypertrophy). This study was undertaken to establish the role of different protein kinase-C (PKC) isoforms in the regulation of cardiac adaptation during two types of cardiac hypertrophy. Phosphorylation of specific PKC-isoforms and expression of their downstream proteins were studied during physiological and pathological hypertrophy in 24 week male Balb/c mice (Mus musculus) models, by reverse transcriptase-PCR, western blot analysis and M-mode echocardiography for cardiac function analysis. PKC-δ was significantly induced during pathological hypertrophy while PKC-α was exclusively activated during physiological hypertrophy in our study. PKC-δ activation during pathological hypertrophy resulted in cardiomyocyte apoptosis leading to compromised cardiac function and on the other hand, activation of PKC-α during physiological hypertrophy promoted cardiomyocyte growth but down regulated cellular apoptotic load resulting in improved cardiac function. Reversal in PKC-isoform with induced activation of PKC-δ and simultaneous inhibition of phospho-PKC-α resulted in an efficient myocardium to deteriorate considerably resulting in compromised cardiac function during physiological hypertrophy via augmentation of apoptotic and fibrotic load. This is the first report where PKC-α and -δ have been shown to play crucial role in cardiac adaptation during physiological and pathological hypertrophy respectively thereby rendering compromised cardiac function to an otherwise efficient heart by conditional reversal of their activation.
Collapse
|