1
|
Li J, Qi Y, Wang J, Dai C, Chen B, Li Y. Trimetazidine Alleviates Postresuscitation Myocardial Dysfunction and Improves 96-Hour Survival in a Ventricular Fibrillation Rat Model. J Am Heart Assoc 2022; 11:e023378. [PMID: 35261264 PMCID: PMC9075307 DOI: 10.1161/jaha.121.023378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022]
Abstract
Background Myocardial dysfunction is a critical cause of post-cardiac arrest hemodynamic instability and circulatory failure that may lead to early mortality after resuscitation. Trimetazidine is a metabolic agent that has been demonstrated to provide protective effects in myocardial ischemia. However, whether trimetazidine protects against postresuscitation myocardial dysfunction is unknown. Methods and Results Cardiopulmonary resuscitation was initiated after 8 minutes of untreated ventricular fibrillation in Sprague-Dawley rats. Animals were randomized to 4 groups immediately after resuscitation (n=15/group): (1) normothermia control (NTC); (2) targeted temperature management; (3) trimetazidine-normothermia; (4) trimetazidine-targeted temperature management. TMZ was administered at a single dose of 10 mg/kg in rats with trimetazidine. The body temperature was maintained at 34.0°C for 2 hours and then rewarmed to 37.5°C in rats with targeted temperature management. Postresuscitation hemodynamics, 96-hours survival, and pathological analysis were assessed. Heart tissues and blood samples of additional rats (n=6/group) undergoing the same experimental procedure were collected to measure myocardial injury, inflammation and oxidative stress-related biomarkers with ELISA-based quantification assays. Compared with normothermia control, tumor necrosis factor-α, and cardiac troponin-I were significantly reduced, whereas the left ventricular ejection fraction and 96-hours survival rates were significantly improved in the 3 experimental groups. Furthermore, inflammation and oxidative stress-related biomarkers together with collagen volume fraction were significantly decreased in rats undergoing postresuscitation interventions. Conclusions Trimetazidine significantly alleviates postresuscitation myocardial dysfunction and improves survival by decreasing oxidative stress and inflammation in a ventricular fibrillation rat model. A single dose of trimetazidine administrated immediately after resuscitation can effectively improve cardiac function, whether used alone or combined with targeted temperature management.
Collapse
Affiliation(s)
- Jingru Li
- Department of Biomedical Engineering and Imaging MedicineArmy Medical UniversityChongqingChina
| | - Yuantong Qi
- Department of PharmaceuticsCollege of PharmacyArmy Medical UniversityChongqingChina
| | - Jianjie Wang
- Department of Biomedical Engineering and Imaging MedicineArmy Medical UniversityChongqingChina
| | - Chenxi Dai
- Department of Biomedical Engineering and Imaging MedicineArmy Medical UniversityChongqingChina
| | - Bihua Chen
- Department of Biomedical Engineering and Imaging MedicineArmy Medical UniversityChongqingChina
| | - Yongqin Li
- Department of Biomedical Engineering and Imaging MedicineArmy Medical UniversityChongqingChina
| |
Collapse
|
2
|
Wu SN, Tsai MS, Huang CH, Chen WJ. Omecamtiv mecarbil treatment improves post-resuscitation cardiac function and neurological outcome in a rat model. PLoS One 2022; 17:e0264165. [PMID: 35176110 PMCID: PMC8853579 DOI: 10.1371/journal.pone.0264165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Background Myocardial dysfunction is a major cause of poor outcomes in the post-cardiac arrest period. Omecamtiv mecarbil (OM) is a selective small molecule activator of cardiac myosin that prolongs myocardial systole and increases stroke volume without apparent effects on myocardial oxygen demand. OM administration is safe and improves cardiac function in patients with acute heart failure. Whether OM improves post-resuscitation myocardial dysfunction remains unclear. This study investigated the effect of OM treatment on post-resuscitation myocardial dysfunction and outcomes. Methods and results Adult male rats were resuscitated after 9.5 min of asphyxia-induced cardiac arrest. OM and normal saline was continuously intravenously infused after return of spontaneous circulation (ROSC) at 0.25 mg/kg/h for 4 h in the experimental group and control group, respectively (n = 20 in each group). Hemodynamic parameters were measured hourly and monitored for 4 h after cardiac arrest. Recovery of neurological function was evaluated by neurological functioning scores (0–12; favorable: 11–12) for rats 72 h after cardiac arrest. OM treatment prolonged left ventricular ejection time and improved post-resuscitation cardiac output. Post-resuscitation heart rate and left ventricular systolic function (dp/dt40) were not different between groups. Kaplan-Meier analysis showed non-statistically higher 72-h survival in the OM group (72.2% [13/18] and 58.8% [10/17], p = 0.386). The OM group had a higher chance of having favorable neurological outcomes in surviving rats 72 h after cardiac arrest (84.6% [11/13] vs. 40% [4/10], p = 0.026). The percentage of damaged neurons was lower in the OM group in a histology study at 72 h after cardiac arrest (55.5±2.3% vs. 76.2±10.2%, p = 0.004). Conclusions OM treatment improved post-resuscitation myocardial dysfunction and neurological outcome in an animal model. These findings support further pre-clinical studies to improve outcomes in post-cardiac arrest care.
Collapse
Affiliation(s)
- Shih-Ni Wu
- Department of Emergency Medicine, National Taiwan University Medical College and Hospital, Taipei, Taiwan
| | - Min-Shan Tsai
- Department of Emergency Medicine, National Taiwan University Medical College and Hospital, Taipei, Taiwan
| | - Chien-Hua Huang
- Department of Emergency Medicine, National Taiwan University Medical College and Hospital, Taipei, Taiwan
- * E-mail:
| | - Wen-Jone Chen
- Department of Emergency Medicine, National Taiwan University Medical College and Hospital, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Medical College and Hospital, Taipei, Taiwan
| |
Collapse
|
3
|
Urocortin Role in Ischemia Cardioprotection and the Adverse Cardiac Remodeling. Int J Mol Sci 2021; 22:ijms222212115. [PMID: 34829997 PMCID: PMC8622004 DOI: 10.3390/ijms222212115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/24/2021] [Accepted: 11/05/2021] [Indexed: 11/17/2022] Open
Abstract
Despite the considerable progress in strategies of myocardial protection, ischemic heart diseases (IHD) and consequent heart failure (HF) remain the main cause of mortality worldwide. Several procedures are used routinely to guarantee the prompt and successful reestablishment of blood flow to preserve the myocardial viability of infarcted hearts from ischemia injuries. However, ischemic heart reperfusion/revascularization triggers additional damages that occur when oxygen-rich blood re-enters the vulnerable myocardial tissue, which is a phenomenon known as ischemia and reperfusion (I/R) syndrome. Complications of I/R injuries provoke the adverse cardiac remodeling, involving inflammation, mishandling of Ca2+ homeostasis, apoptotic genes activation, cardiac myocytes loss, etc., which often progress toward HF. Therefore, there is an urgent need to develop new cardioprotective therapies for IHD and HF. Compelling evidence from animal studies and pilot clinical trials in HF patients suggest that urocortin (Ucn) isoforms, which are peptides associated with stress and belonging to the corticotropin releasing factor family, have promising potential to improve cardiovascular functions by targeting many signaling pathways at different molecular levels. This review highlights the current knowledge on the role of urocortin isoforms in cardioprotection, focusing on its acute and long-term effects.
Collapse
|
4
|
Popov SV, Prokudina ES, Mukhomedzyanov AV, Naryzhnaya NV, Ma H, Zurmanova JM, der Ven PFMV, Maslov LN. Cardioprotective and Vasoprotective Effects of Corticotropin-Releasing Hormone and Urocortins: Receptors and Signaling. J Cardiovasc Pharmacol Ther 2021; 26:575-584. [PMID: 34351805 DOI: 10.1177/1074248420985301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite the recent progress in research and therapy, cardiovascular diseases are still the most common cause of death worldwide, thus new approaches are still needed. The aim of this review is to highlight the cardioprotective potential of urocortins and corticotropin-releasing hormone (CRH) and their signaling. It has been documented that urocortins and CRH reduce ischemic and reperfusion (I/R) injury, prevent reperfusion ventricular tachycardia and fibrillation, and improve cardiac contractility during reperfusion. Urocortin-induced increase in cardiac tolerance to I/R depends mainly on the activation of corticotropin-releasing hormone receptor-2 (CRHR2) and its downstream pathways including tyrosine kinase Src, protein kinase A and C (PKA, PKCε) and extracellular signal-regulated kinase (ERK1/2). It was discussed the possibility of the involvement of interleukin-6, Janus kinase-2 and signal transducer and activator of transcription 3 (STAT3) and microRNAs in the cardioprotective effect of urocortins. Additionally, phospholipase-A2 inhibition, mitochondrial permeability transition pore (MPT-pore) blockade and suppression of apoptosis are involved in urocortin-elicited cardioprotection. Chronic administration of urocortin-2 prevents the development of postinfarction cardiac remodeling. Urocortin possesses vasoprotective and vasodilator effect; the former is mediated by PKC activation and prevents an impairment of endothelium-dependent coronary vasodilation after I/R in the isolated heart, while the latter includes both cAMP and cGMP signaling and its downstream targets. As CRHR2 is expressed by both cardiomyocytes and vascular endothelial cells. Urocortins mediate both endothelium-dependent and -independent relaxation of coronary arteries.
Collapse
Affiliation(s)
- Sergey V Popov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Ekaterina S Prokudina
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Alexander V Mukhomedzyanov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Natalia V Naryzhnaya
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Huijie Ma
- Department of Physiology, 12553Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jitka M Zurmanova
- Department of Physiology, Faculty of Science, 37740Charles University, Prague, Czech Republic
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, 9374University of Bonn, Bonn, Germany
| | - Leonid N Maslov
- Laboratory of Experimental Cardiology, 164253Cardiology Research Institute, Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| |
Collapse
|
5
|
Salvianolic Acid B Improves Postresuscitation Myocardial and Cerebral Outcomes in a Murine Model of Cardiac Arrest: Involvement of Nrf2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1605456. [PMID: 32714485 PMCID: PMC7352143 DOI: 10.1155/2020/1605456] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023]
Abstract
Survival and outcome of cardiac arrest (CA) are dismal despite improvements in cardiopulmonary resuscitation (CPR). Salvianolic acid B (Sal B), extracted from Salvia miltiorrhiza, has been investigated for its cardioprotective properties in cardiac remodeling and ischemic heart disease, but less is known about its role in CA. The aim of this study was to learn whether Sal B improves cardiac and neurologic outcomes after CA/CPR in mice. Female C57BL/6 mice were subjected to eight minutes of CA induced by an intravenous injection of potassium chloride (KCl), followed by CPR. After 30 seconds of CPR, mice were blindly randomized to receive either Sal B (20 mg/kg) or vehicle (normal saline) intravenously. Hemodynamic variables and indices of left ventricular function were determined before CA and within three hours after CPR, the early postresuscitation period. Sal B administration resulted in a remarkable decrease in the time required for the return of spontaneous circulation (ROSC) in animals that successfully resuscitated compared to the vehicle-treated mice. Myocardial performance, including cardiac output and left ventricular systolic (dp/dtmax) and diastolic (dp/dtmin) function, was clearly ameliorated within three hours of ROSC in the Sal B-treated mice. Moreover, Sal B inhibited CA/CPR-induced cardiomyocyte apoptosis and preserved mitochondrial morphology and function. Mechanistically, Sal B dramatically promoted Nrf2 nuclear translocation through the downregulation of Keap1, which resulted in the expression of antioxidant enzymes, including HO-1 and NQO1, thereby counteracted the oxidative damage in response to CA/CPR. The aforementioned antiapoptotic and antioxidant effects of Sal B were impaired in the setting of gene silencing of Nrf2 with siRNA in vitro model. These improvements were associated with better neurological function and increased survival rate (75% vs. 40%, p < 0.05) up to 72 hours postresuscitation. Our findings suggest that the administration of Sal B improved cardiac function and neurological outcomes in a murine model of CA via activating the Nrf2 antioxidant signaling pathway, which may represent a novel therapeutic strategy for the treatment of CA.
Collapse
|
6
|
Wang CH, Chang WT, Huang CH, Tsai MS, Liu SH, Chen WJ. Cerebral Blood Flow-Guided Manipulation of Arterial Blood Pressure Attenuates Hippocampal Apoptosis After Asphyxia-Induced Cardiac Arrest in Rats. J Am Heart Assoc 2020; 9:e016513. [PMID: 32552439 PMCID: PMC7670514 DOI: 10.1161/jaha.120.016513] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background In most post-cardiac arrest patients, the autoregulation mechanism of cerebral blood flow (CBF) is dysregulated. We examined whether recovery of CBF by adjusting mean arterial pressure mitigates post-cardiac arrest neuronal damage. Methods and Results Wistar rats that underwent 8-minute asphyxia-induced cardiac arrest and resuscitation were computer-randomized to norepinephrine or control groups. The CBF was measured at the dorsal hippocampal CA1 region of the left hemisphere. In the norepinephrine group, the mean arterial pressure was adjusted to recover CBF to 80% to 100% of baseline. Twenty-four hours following resuscitation, neurological outcomes were assessed, and brain tissues and blood samples were harvested for neuronal apoptosis and injury assessment. Thirty resuscitated rats were randomized into 2 groups, each containing 12 rats that completed the experiments. Norepinephrine infusion effectively prevented posthyperemia hypoperfusion and recovered CBF to pre-arrest baseline levels; a moderate positive linear correlation between mean arterial pressure and CBF during this period was also observed (P<0.001). There were no significant between-group differences in neurological recovery. In the norepinephrine group compared with the control group, upregulated cleaved caspase-3 protein expression in brain tissue determined by Western blot was reduced (P=0.02) and the densities of apoptotic cells in hippocampal CA1 and CA3 regions determined by terminal deoxynucleotidyl transferase-mediated dUTP biotin nick-end labeling were decreased (P<0.001). No significant differences in serum neuron-specific enolase or S100β levels were detected between the 2 groups. Conclusions CBF recovery demonstrated neuroprotective effects by reducing activation of cerebral apoptosis and number of apoptotic neurons. However, these effects did not significantly improve clinical neurological function, necessitating further investigation.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Wei-Tien Chang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Chien-Hua Huang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Min-Shan Tsai
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology College of Medicine National Taiwan University Taipei Taiwan.,Department of Medical Research China Medical University Hospital China Medical University Taichung Taiwan.,Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
| | - Wen-Jone Chen
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan.,Division of Cardiology Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| |
Collapse
|
7
|
Wang CH, Chang WT, Tsai MS, Huang CH, Chen WJ. Synergistic Effects of Moderate Therapeutic Hypothermia and Levosimendan on Cardiac Function and Survival After Asphyxia-Induced Cardiac Arrest in Rats. J Am Heart Assoc 2020; 9:e016139. [PMID: 32476598 PMCID: PMC7429058 DOI: 10.1161/jaha.120.016139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background This study investigated whether levosimendan, an inotropic calcium sensitizer, when combined with moderate therapeutic hypothermia, may exert synergistic benefits on post–cardiac arrest myocardial dysfunction and improve outcomes. Methods and Results After 9.5‐minute asphyxia‐induced cardiac arrest and resuscitation, 48 rats were randomized equally into 4 groups following return of spontaneous circulation (ROSC), including normothermia, hypothermia, normothermia–levosimendan, and hypothermia–levosimendan groups. For the normothermia group, the target temperature was 37°C while for the hypothermia group, the target temperature was 32°C, both of which were to be maintained for 4 hours after ROSC. Levosimendan was administered after ROSC with a loading dose of 10 μg/kg and then infused at 0.1 μg/kg per min for 4 hours. In the hypothermia–levosimendan group, left ventricular systolic function and cardiac output increased significantly, whereas the heart rate and systemic vascular resistance decreased significantly compared with the normothermia group. Also, the concentrations of interleukin 1β at 4 hours post‐ROSC and the production of NO between 1 hour and 4 hours post‐ROSC were reduced significantly in the hypothermia–levosimendan group compared with the normothermia group. The 72‐hour post‐ROSC survival and neurological recovery were also significantly better in the hypothermia–levosimendan group compared with the normothermia group (survival, 100% versus 50%, χ2 test, P=0.006). Conclusions Compared with normothermia, only combined moderate therapeutic hypothermia and levosimendan treatment could consistently improve post–cardiac arrest myocardial dysfunction and decrease the release of pro‐inflammatory molecules, thereby improving survival and neurological outcomes. These findings suggest synergistic benefits between moderate therapeutic hypothermia and levosimendan.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Wei-Tien Chang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Min-Shan Tsai
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Chien-Hua Huang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Wen-Jone Chen
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan.,Division of Cardiology Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| |
Collapse
|
8
|
Grisanti LA, Schumacher SM, Tilley DG, Koch WJ. Designer Approaches for G Protein-Coupled Receptor Modulation for Cardiovascular Disease. JACC Basic Transl Sci 2018; 3:550-562. [PMID: 30175279 PMCID: PMC6115700 DOI: 10.1016/j.jacbts.2017.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022]
Abstract
The new horizon for cardiac therapy may lie beneath the surface, with the downstream mediators of G protein–coupled receptor (GPCR) activity. Targeted approaches have shown that receptor activation may be biased toward signaling through G proteins or through GPCR kinases (GRKs) and β-arrestins, with divergent functional outcomes. In addition to these canonical roles, numerous noncanonical activities of GRKs and β-arrestins have been demonstrated to modulate GPCR signaling at all levels of receptor activation and regulation. Further, research continues to identify novel GRK/effector and β-arrestin/effector complexes with distinct impacts on cardiac function in the normal heart and the diseased heart. Coupled with the identification of once orphan receptors and endogenous ligands with beneficial cardiovascular effects, this expands the repertoire of GPCR targets. Together, this research highlights the potential for focused therapeutic activation of beneficial pathways, with simultaneous exclusion or inhibition of detrimental signaling, and represents a new wave of therapeutic development.
Collapse
Key Words
- AR, adrenergic receptor
- AT1R, angiotensin II type 1A receptor
- CRF, corticotropin-releasing factor
- EGFR, epidermal growth factor receptor
- ERK1/2, extracellular signal-regulated kinase
- G protein–coupled receptor kinases
- G protein–coupled receptors
- GPCR, G protein–coupled receptor
- GRK, G protein–coupled receptor kinase
- HF, heart failure
- ICL, intracellular loop
- PI3K, phosphoinositide 3-kinase
- SERCA2a, sarco(endo)plasmic reticulum Ca2+-ATPase
- SII, [Sar(1), Ile (4), Ile(8)]-angiotensin II
- biased ligands
Collapse
Affiliation(s)
- Laurel A Grisanti
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Sarah M Schumacher
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Douglas G Tilley
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|