51
|
Kant S, Kesarwani P, Guastella AR, Kumar P, Graham SF, Buelow KL, Nakano I, Chinnaiyan P. Perhexiline Demonstrates FYN-mediated Antitumor Activity in Glioblastoma. Mol Cancer Ther 2020; 19:1415-1422. [PMID: 32430486 DOI: 10.1158/1535-7163.mct-19-1047] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/16/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022]
Abstract
Glioblastoma is the most common primary malignant brain tumor in adults. Despite aggressive treatment, outcomes remain poor with few long-term survivors. Therefore, considerable effort is being made to identify novel therapies for this malignancy. Targeting tumor metabolism represents a promising therapeutic strategy and activation of fatty acid oxidation (FAO) has been identified as a central metabolic node contributing toward gliomagenesis. Perhexiline is a compound with a long clinical track record in angina treatment and commonly described as an FAO inhibitor. We therefore sought to determine whether this compound might be repurposed to serve as a novel therapy in glioblastoma. Perhexiline demonstrated potent in vitro cytotoxicity, induction of redox stress and apoptosis in a panel of glioblastoma cell lines. However, the antitumor activity of perhexiline was distinct when compared with the established FAO inhibitor etomoxir. By evaluating mitochondrial respiration and lipid dynamics in glioblastoma cells following treatment with perhexiline, we confirmed this compound did not inhibit FAO in our models. Using in silico approaches, we identified FYN as a probable target of perhexiline and validated the role of this protein in perhexiline sensitivity. We extended studies to patient samples, validating the potential of FYN to serve as therapeutic target in glioma. When evaluated in vivo, perhexiline demonstrated the capacity to cross the blood-brain barrier and antitumor activity in both flank and orthotopic glioblastoma models. Collectively, we identified potent FYN-dependent antitumor activity of perhexiline in glioblastoma, thereby, representing a promising agent to be repurposed for the treatment of this devastating malignancy.
Collapse
Affiliation(s)
- Shiva Kant
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - Pravin Kesarwani
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | | | - Praveen Kumar
- Metabolomics and Obstetrics/Gynecology, Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan
| | - Stewart F Graham
- Metabolomics and Obstetrics/Gynecology, Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan
| | - Katie L Buelow
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - Ichiro Nakano
- Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prakash Chinnaiyan
- Department of Radiation Oncology, Beaumont Health, Royal Oak, Michigan. .,Oakland University William Beaumont School of Medicine, Royal Oak, Michigan
| |
Collapse
|
52
|
Nallanthighal S, Rada M, Heiserman JP, Cha J, Sage J, Zhou B, Yang W, Hu Y, Korgaonkar C, Hanos CT, Ashkavand Z, Norman K, Orsulic S, Cheon DJ. Inhibition of collagen XI alpha 1-induced fatty acid oxidation triggers apoptotic cell death in cisplatin-resistant ovarian cancer. Cell Death Dis 2020; 11:258. [PMID: 32312965 PMCID: PMC7171147 DOI: 10.1038/s41419-020-2442-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/12/2022]
Abstract
Collagen type XI alpha 1 (COL11A1) is a novel biomarker associated with cisplatin resistance in ovarian cancer. However, the mechanisms underlying how COL11A1 confers cisplatin resistance in ovarian cancer are poorly understood. We identified that fatty acid β-oxidation (FAO) is upregulated by COL11A1 in ovarian cancer cells and that COL11A1-driven cisplatin resistance can be abrogated by inhibition of FAO. Furthermore, our results demonstrate that COL11A1 also enhances the expression of proteins involved in fatty acid synthesis. Interestingly, COL11A1-induced upregulation of fatty acid synthesis and FAO is modulated by the same signaling molecules. We identified that binding of COL11A1 to its receptors, α1β1 integrin and discoidin domain receptor 2 (DDR2), activates Src-Akt-AMPK signaling to increase the expression of both fatty acid synthesis and oxidation enzymes, although DDR2 seems to be the predominant receptor. Inhibition of fatty acid synthesis downregulates FAO despite the presence of COL11A1, suggesting that fatty acid synthesis might be a driver of FAO in ovarian cancer cells. Taken together, our results suggest that COL11A1 upregulates fatty acid metabolism in ovarian cancer cells in a DDR2-Src-Akt-AMPK dependent manner. Therefore, we propose that blocking FAO might serve as a promising therapeutic target to treat ovarian cancer, particularly cisplatin-resistant recurrent ovarian cancers which typically express high levels of COL11A1.
Collapse
Affiliation(s)
- Sameera Nallanthighal
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Miran Rada
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - James Patrick Heiserman
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Jennifer Cha
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Jessica Sage
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Bo Zhou
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Wei Yang
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ye Hu
- Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedar-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Chaitali Korgaonkar
- Department of Obstetrics and Gynecology, Albany Medical College, Albany, NY, 12208, USA
| | | | - Zahra Ashkavand
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Kenneth Norman
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Sandra Orsulic
- Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedar-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dong-Joo Cheon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
53
|
Maltês S, Lopes LR. New perspectives in the pharmacological treatment of hypertrophic cardiomyopathy. Rev Port Cardiol 2020; 39:99-109. [PMID: 32245685 DOI: 10.1016/j.repc.2019.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/17/2019] [Accepted: 03/10/2019] [Indexed: 10/24/2022] Open
Abstract
Hypertrophic cardiomyopathy is an inherited cardiac disease and a major cause of heart failure and sudden death. Even though it was described more than 50 years ago, sarcomeric hypertrophic cardiomyopathy still lacks a disease-specific treatment. The drugs routinely used alleviate symptoms but do not prevent or revert the phenotype. With recent advances in the knowledge about the genetics and pathophysiology of hypertrophic cardiomyopathy, new genetic and pharmacological approaches have been recently discovered and studied that, by influencing different pathways involved in this disease, have the potential to function as disease-modifying therapies. These promising new pharmacological and genetic therapies will be the focus of this review.
Collapse
Affiliation(s)
- Sérgio Maltês
- Clínica Universitária de Cardiologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal.
| | - Luis Rocha Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, Inglaterra; St. Bartholomew's Hospital, Barts Heart Centre, London, Inglaterra; Centro Cardiovascular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
54
|
Murabito A, Hirsch E, Ghigo A. Mechanisms of Anthracycline-Induced Cardiotoxicity: Is Mitochondrial Dysfunction the Answer? Front Cardiovasc Med 2020; 7:35. [PMID: 32226791 PMCID: PMC7080657 DOI: 10.3389/fcvm.2020.00035] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac side effects are a major drawback of anticancer therapies, often requiring the use of low and less effective doses or even discontinuation of the drug. Among all the drugs known to cause severe cardiotoxicity are anthracyclines that, though being the oldest chemotherapeutic drugs, are still a mainstay in the treatment of solid and hematological tumors. The recent expansion of the field of Cardio-Oncology, a branch of cardiology dealing with prevention or treatment of heart complications due to cancer treatment, has greatly improved our knowledge of the molecular mechanisms behind anthracycline-induced cardiotoxicity (AIC). Despite excessive generation of reactive oxygen species was originally believed to be the main cause of AIC, recent evidence points to the involvement of a plethora of different mechanisms that, interestingly, mainly converge on deregulation of mitochondrial function. In this review, we will describe how anthracyclines affect cardiac mitochondria and how these organelles contribute to AIC. Furthermore, we will discuss how drugs specifically targeting mitochondrial dysfunction and/or mitochondria-targeted drugs could be therapeutically exploited to treat AIC.
Collapse
Affiliation(s)
- Alessandra Murabito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
55
|
Bairey Merz CN, Pepine CJ, Shimokawa H, Berry C. Treatment of coronary microvascular dysfunction. Cardiovasc Res 2020; 116:856-870. [PMID: 32087007 PMCID: PMC7061279 DOI: 10.1093/cvr/cvaa006] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/18/2019] [Indexed: 12/30/2022] Open
Abstract
Contemporary data indicate that patients with signs and symptoms of ischaemia and non-obstructive coronary artery disease (INOCA) often have coronary microvascular dysfunction (CMD) with elevated risk for adverse outcomes. Coronary endothelial (constriction with acetylcholine) and/or microvascular (limited coronary flow reserve with adenosine) dysfunction are well-documented, and extensive non-obstructive atherosclerosis is often present. Despite these data, patients with INOCA currently remain under-treated, in part, because existing management guidelines do not address this large, mostly female population due to the absence of evidence-based data. Relatively small sample-sized, short-term pilot studies of symptomatic mostly women, with INOCA, using intense medical therapies targeting endothelial, microvascular, and/or atherosclerosis mechanisms suggest symptom, ischaemia, and coronary vascular functional improvement, however, randomized, controlled outcome trials testing treatment strategies have not been completed. We review evidence regarding CMD pharmacotherapy. Potent statins in combination with angiotensin-converting enzyme inhibitor (ACE-I) or receptor blockers if intolerant, at maximally tolerated doses appear to improve angina, stress testing, myocardial perfusion, coronary endothelial function, and microvascular function. The Coronary Microvascular Angina trial supports invasive diagnostic testing with stratified therapy as an approach to improve symptoms and quality of life. The WARRIOR trial is testing intense medical therapy of high-intensity statin, maximally tolerated ACE-I plus aspirin on longer-term outcomes to provide evidence for guidelines. Novel treatments and those under development appear promising as the basis for future trial planning.
Collapse
Affiliation(s)
- C Noel Bairey Merz
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai, 127 S. San Vicente Blvd, Suite A3600, Los Angeles, CA 90048, USA
| | - Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida, 1329 SW 16th Street, PO Box 100288, Gainesville, FL 32610-0288, USA
| | - Hiroki Shimokawa
- Division of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Colin Berry
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
56
|
Maltês S, Lopes LR. New perspectives in the pharmacological treatment of hypertrophic cardiomyopathy. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2020. [DOI: 10.1016/j.repce.2019.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
57
|
Schlaepfer IR, Joshi M. CPT1A-mediated Fat Oxidation, Mechanisms, and Therapeutic Potential. Endocrinology 2020; 161:5695911. [PMID: 31900483 DOI: 10.1210/endocr/bqz046] [Citation(s) in RCA: 405] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022]
Abstract
Energy homeostasis during fasting or prolonged exercise depends on mitochondrial fatty acid oxidation (FAO). This pathway is crucial in many tissues with high energy demand and its disruption results in inborn FAO deficiencies. More than 15 FAO genetic defects have been currently described, and pathological variants described in circumpolar populations provide insights into its critical role in metabolism. The use of fatty acids as energy requires more than 2 dozen enzymes and transport proteins, which are involved in the activation and transport of fatty acids into the mitochondria. As the key rate-limiting enzyme of FAO, carnitine palmitoyltransferase I (CPT1) regulates FAO and facilitates adaptation to the environment, both in health and in disease, including cancer. The CPT1 family of proteins contains 3 isoforms: CPT1A, CPT1B, and CPT1C. This review focuses on CPT1A, the liver isoform that catalyzes the rate-limiting step of converting acyl-coenzyme As into acyl-carnitines, which can then cross membranes to get into the mitochondria. The regulation of CPT1A is complex and has several layers that involve genetic, epigenetic, physiological, and nutritional modulators. It is ubiquitously expressed in the body and associated with dire consequences linked with genetic mutations, metabolic disorders, and cancers. This makes CPT1A an attractive target for therapeutic interventions. This review discusses our current understanding of CPT1A expression, its role in heath and disease, and the potential for therapeutic opportunities targeting this enzyme.
Collapse
Affiliation(s)
- Isabel R Schlaepfer
- University of Colorado School of Medicine, Division of Medical Oncology, Aurora
| | - Molishree Joshi
- University of Colorado School of Medicine, Department of Pharmacology, Aurora, Colorado
| |
Collapse
|
58
|
Ma Y, Wang W, Devarakonda T, Zhou H, Wang XY, Salloum FN, Spiegel S, Fang X. Functional analysis of molecular and pharmacological modulators of mitochondrial fatty acid oxidation. Sci Rep 2020; 10:1450. [PMID: 31996743 PMCID: PMC6989517 DOI: 10.1038/s41598-020-58334-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/13/2020] [Indexed: 01/02/2023] Open
Abstract
Fatty acid oxidation (FAO) is a key bioenergetic pathway often dysregulated in diseases. The current knowledge on FAO regulators in mammalian cells is limited and sometimes controversial. Previous FAO analyses involve nonphysiological culture conditions or lack adequate quantification. We herein described a convenient and quantitative assay to monitor dynamic FAO activities of mammalian cells in physiologically relevant settings. The method enabled us to assess various molecular and pharmacological modulators of the FAO pathway in established cell lines, primary cells and mice. Surprisingly, many previously proposed FAO inhibitors such as ranolazine and trimetazidine lacked FAO-interfering activity. In comparison, etomoxir at low micromolar concentrations was sufficient to saturate its target proteins and to block cellular FAO function. Oxfenicine, on the other hand, acted as a partial inhibitor of FAO. As another class of FAO inhibitors that transcriptionally repress FAO genes, antagonists of peroxisome proliferator-activated receptors (PPARs), particularly that of PPARα, significantly decreased cellular FAO activity. Our assay also had sufficient sensitivity to monitor upregulation of FAO in response to environmental glucose depletion and other energy-demanding cues. Altogether this study provided a reliable FAO assay and a clear picture of biological properties of potential FAO modulators in the mammalian system.
Collapse
Affiliation(s)
- Yibao Ma
- Departments of Biochemistry & Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Wei Wang
- Departments of Biochemistry & Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Teja Devarakonda
- Internal Medicine/Cardiology Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Huiping Zhou
- Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Xiang-Yang Wang
- Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Fadi N Salloum
- Internal Medicine/Cardiology Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Sarah Spiegel
- Departments of Biochemistry & Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA
| | - Xianjun Fang
- Departments of Biochemistry & Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, 23298, USA.
| |
Collapse
|
59
|
Mhaldar SN, Tilve SG. Total Synthesis of 2‐(2‐Ketoalkyl)‐piperidine Alkaloids (+)‐ Pelletierine, (+)‐Haloxynine and (‐)‐ N‐Methyl‐pelletierine via Regioselective Wacker Oxidation. ChemistrySelect 2019. [DOI: 10.1002/slct.201902115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Shashank N. Mhaldar
- School of Chemical SciencesGoa University, Taleigao Plateau Goa 403206 India
| | - Santosh G. Tilve
- School of Chemical SciencesGoa University, Taleigao Plateau Goa 403206 India
| |
Collapse
|
60
|
Hu W, Lu H, Zhang J, Fan Y, Chang Z, Liang W, Wang H, Zhu T, Garcia-Barrio MT, Peng D, Chen YE, Guo Y. Krüppel-like factor 14, a coronary artery disease associated transcription factor, inhibits endothelial inflammation via NF-κB signaling pathway. Atherosclerosis 2018; 278:39-48. [PMID: 30248551 PMCID: PMC6441279 DOI: 10.1016/j.atherosclerosis.2018.09.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/19/2018] [Accepted: 09/14/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS Human genetic studies indicated that variations near the transcription factor Krüppel-like factor 14 (KLF14) gene locus are highly associated with coronary artery disease. Activation of endothelial cells (ECs) by pro-inflammatory molecules and pathways is a primary step in atherosclerosis development. We aimed to investigate the effects and mechanism of KLF14 on inflammatory responses in ECs. METHODS Adenovirus-mediated overexpression of human KLF14 and EC specific Klf14 knockout mice were applied to study the role of KLF14 in EC inflammation. Intravital microscopy was used to examine leukocyte-endothelial cell interactions in vivo. RESULTS The expression of Klf14 was markedly decreased in mouse aortic ECs in both acute and chronic inflammatory conditions. Overexpression of KLF14 inhibited inflammatory activation of human ECs stimulated by interleukin 1β and tumor necrosis factor α. Primary pulmonary ECs from Klf14 knockout mice showed increased expression of adhesion molecules under IL-1β stimuli. Mechanistically, KLF14 inhibited NF-κB signaling pathway by transcriptionally suppressing the expression of p65, resulting in significantly decreased leukocyte adhesion to activated ECs. Using intravital microscopy, an increased leukocyte-endothelial cell interaction was observed in endothelial specific Klf14 knockout mice compared to wild type control mice. Additionally, perhexiline, a KLF14 activator, induces KLF14 expression in ECs and reduced leukocyte-endothelial cell interactions in vitro and in vivo. CONCLUSIONS The data revealed that KLF14 inhibited the inflammatory response in ECs and the protective effects were mediated by transcriptional inhibition of NF-κB signaling pathway. Endothelial KLF14 could be a potential therapeutic target for cardiovascular diseases.
Collapse
Affiliation(s)
- Wenting Hu
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA; Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Haocheng Lu
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Jifeng Zhang
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yanbo Fan
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Ziyi Chang
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Wenying Liang
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Huilun Wang
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Tianqing Zhu
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Minerva T Garcia-Barrio
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Y Eugene Chen
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| | - Yanhong Guo
- From Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
61
|
|
62
|
Kamalian L, Douglas O, Jolly CE, Snoeys J, Simic D, Monshouwer M, Williams DP, Kevin Park B, Chadwick AE. The utility of HepaRG cells for bioenergetic investigation and detection of drug-induced mitochondrial toxicity. Toxicol In Vitro 2018; 53:136-147. [PMID: 30096366 DOI: 10.1016/j.tiv.2018.08.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/03/2018] [Accepted: 08/05/2018] [Indexed: 12/12/2022]
Abstract
The importance of mitochondrial toxicity in drug-induced liver injury is well established. The bioenergetic phenotype of the HepaRG cell line was defined in order to assess their suitability as a model of mitochondrial hepatotoxicity. Bioenergetic phenotyping categorised the HepaRG cells as less metabolically active when measured beside the more energetic HepG2 cells. However, inhibition of mitochondrial ATP synthase induced an increase in glycolytic activity of both HepaRG and HepG2 cells suggesting an active Crabtree Effect in both cell lines. The suitability of HepaRG cells for the acute metabolic modification assay as a screen for mitotoxicity was confirmed using a panel of compounds, including both positive and negative mitotoxic compounds. Seahorse respirometry studies demonstrated that a statistically significant decrease in spare respiratory capacity is the first indication of mitochondrial dysfunction. Furthermore, based upon comparing changes in respiratory parameters to those of the positive controls, rotenone and carbonyl cyanide m-chlorophenyl hydrazone, compounds were categorised into two mechanistic groups; inhibitors or uncouplers of the electron transport chain. Overall, the findings from this study have demonstrated that HepaRG cells, despite having different resting bioenergetic phenotype to HepG2 cells are a suitable model to detect drug-induced mitochondrial toxicity with similar detection rates to HepG2 cells.
Collapse
Affiliation(s)
- Laleh Kamalian
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| | - Oisin Douglas
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Carol E Jolly
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| | - Jan Snoeys
- Pharmacokinetics Dynamics and Metabolism, Janssen Research and Development, Beerse, Belgium.
| | - Damir Simic
- Mechanistic and Investigative Toxicology, Janssen Research and Development, Spring House, PA, USA.
| | - Mario Monshouwer
- Pharmacokinetics Dynamics and Metabolism, Janssen Research and Development, Beerse, Belgium
| | - Dominic P Williams
- Innovative Medicines and Early Development
- Drug Safety and Metabolism
- Translational Safety, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge, CB4 0FZ, United Kingdom.
| | - B Kevin Park
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| | - Amy E Chadwick
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| |
Collapse
|
63
|
Draoui N, de Zeeuw P, Carmeliet P. Angiogenesis revisited from a metabolic perspective: role and therapeutic implications of endothelial cell metabolism. Open Biol 2018; 7:rsob.170219. [PMID: 29263247 PMCID: PMC5746547 DOI: 10.1098/rsob.170219] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/28/2017] [Indexed: 12/16/2022] Open
Abstract
Endothelial cell (EC) metabolism has lately emerged as a novel and promising therapeutic target to block vascular dysregulation associated with diseases like cancer and blinding eye disease. Glycolysis, fatty acid oxidation (FAO) and, more recently, glutamine/asparagine metabolism emerged as key regulators of EC metabolism, able to impact angiogenesis in health and disease. ECs are highly glycolytic as they require ATP and biomass for vessel sprouting. Notably, a regulator of the glycolytic pathway, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3, controls vessel sprouting during the angiogenic switch and its inhibition in tumour ECs leads to vessel normalization, thereby reducing metastasis and ameliorating chemotherapy. Moreover, FAO promotes EC proliferation through DNA synthesis, and plays an essential role in lymphangiogenesis via epigenetic regulation of histone acetylation. Pathological angiogenesis was decreased upon blockade of carnitine palmitoyltransferase 1, a regulator of FAO in ECs. More recently, metabolism of glutamine, in conjunction with asparagine, was reported to maintain EC sprouting through TCA anaplerosis, redox homeostasis, mTOR activation and endoplasmic stress control. Inactivation or blockade of glutaminase 1, which hydrolyses glutamine into ammonia and glutamate, impairs angiogenesis in health and disease, while silencing of asparagine synthetase reduces vessel sprouting in vitro. In this review, we summarize recent insights into EC metabolism and discuss therapeutic implications of targeting EC metabolism.
Collapse
Affiliation(s)
- Nihed Draoui
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, KU Leuven, Campus Gasthuisberg O&N4, Herestraat 49-912, Leuven 3000, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, KU Leuven, Campus Gasthuisberg O&N4, Herestraat 49-912, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium .,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, KU Leuven, Campus Gasthuisberg O&N4, Herestraat 49-912, Leuven 3000, Belgium
| |
Collapse
|
64
|
Tyagi R, Maddirala AR, Elfawal M, Fischer C, Bulman CA, Rosa BA, Gao X, Chugani R, Zhou M, Helander J, Brindley PJ, Tseng CC, Greig IR, Sakanari J, Wildman SA, Aroian R, Janetka JW, Mitreva M. Small Molecule Inhibitors of Metabolic Enzymes Repurposed as a New Class of Anthelmintics. ACS Infect Dis 2018; 4:1130-1145. [PMID: 29718656 PMCID: PMC6283408 DOI: 10.1021/acsinfecdis.8b00090] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The enormous prevalence of infections caused by parasitic nematodes worldwide, coupled to the rapid emergence of their resistance to commonly used anthelmintic drugs, presents an urgent need for the discovery of new drugs. Herein, we have identified several classes of small molecules with broad spectrum activity against these pathogens. Previously, we reported the identification of carnitine palmitoyltransferases (CPTs) as a representative class of enzymes as potential targets for metabolic chokepoint intervention that was elucidated from a combination of chemogenomic screening and experimental testing in nematodes. Expanding on these previous findings, we have discovered that several chemical classes of known small molecule inhibitors of mammalian CPTs have potent activity as anthelmintics. Cross-clade efficacy against a broad spectrum of adult parasitic nematodes was demonstrated for multiple compounds from different series. Several analogs of these initial hit compounds were designed and synthesized. The compounds we report represent a good starting point for further lead identification and optimization for development of new anthelmintic drugs with broad spectrum activity and a novel mechanism of action.
Collapse
Affiliation(s)
- Rahul Tyagi
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
| | - Amarendar Reddy Maddirala
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Mostafa Elfawal
- University of Massachusetts Medical School, Suite 219 Biotech 2, 373 Plantation St., Worcester, Massachusetts 01605, USA
| | - Chelsea Fischer
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th St, San Francisco, California 94158, USA
| | - Christina A. Bulman
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th St, San Francisco, California 94158, USA
| | - Bruce A. Rosa
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
| | - Xin Gao
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
| | - Ryan Chugani
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Mingzhou Zhou
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Jon Helander
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Paul J. Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, Ross Hall, Room 521, 2300 Eye Street, NW, Washington, DC 20037, USA
| | - Chih-Chung Tseng
- Kosterlitz Centre for Therapeutics, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, U.K
| | - Iain R. Greig
- Kosterlitz Centre for Therapeutics, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, U.K
| | - Judy Sakanari
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th St, San Francisco, California 94158, USA
| | - Scott A. Wildman
- UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53792, USA
| | - Raffi Aroian
- University of Massachusetts Medical School, Suite 219 Biotech 2, 373 Plantation St., Worcester, Massachusetts 01605, USA
| | - James W. Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, 4523 Clayton Ave., CB 8051, St. Louis MO, 63110, USA
| |
Collapse
|
65
|
Beezhold K, Byersdorfer CA. Targeting immuno-metabolism to improve anti-cancer therapies. Cancer Lett 2017; 414:127-135. [PMID: 29126914 DOI: 10.1016/j.canlet.2017.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/23/2017] [Accepted: 11/04/2017] [Indexed: 01/07/2023]
Abstract
The immunology community has made significant strides in recent years in using the immune system to target and eliminate cancer. Therapies such as hematopoietic stem cell transplantation (HSCT) are the standard of care treatment for several malignancies, while therapies incorporating chimeric antigen receptor (CAR) T cells or checkpoint molecule blockade have been revolutionary. However, these approaches are not optimal for all cancers and in some cases, have failed outright. The greatest obstacle to making these therapies more effective may be rooted in one of the most basic concepts of cell biology, metabolism. Research over the last decade has revealed that T cell proliferation and differentiation is intimately linked to robust changes in metabolic activity, delineation of which may provide ways to manipulate the immuno-oncologic responses to our advantage. Here, we provide a basic overview of T cell metabolism, discuss what is known about metabolic regulation of T cells during allogeneic HSCT, point to evidence on the importance of T cell metabolism during CAR T cell and solid tumor therapies, and speculate about the role for compounds that might have dual-action on both immune cells and tumor cells simultaneously.
Collapse
Affiliation(s)
- Kevin Beezhold
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Craig A Byersdorfer
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| |
Collapse
|
66
|
Noordali H, Loudon BL, Frenneaux MP, Madhani M. Cardiac metabolism - A promising therapeutic target for heart failure. Pharmacol Ther 2017; 182:95-114. [PMID: 28821397 DOI: 10.1016/j.pharmthera.2017.08.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Both heart failure with reduced ejection fraction (HFrEF) and with preserved ejection fraction (HFpEF) are associated with high morbidity and mortality. Although many established pharmacological interventions exist for HFrEF, hospitalization and death rates remain high, and for those with HFpEF (approximately half of all heart failure patients), there are no effective therapies. Recently, the role of impaired cardiac energetic status in heart failure has gained increasing recognition with the identification of reduced capacity for both fatty acid and carbohydrate oxidation, impaired function of the electron transport chain, reduced capacity to transfer ATP to the cytosol, and inefficient utilization of the energy produced. These nodes in the genesis of cardiac energetic impairment provide potential therapeutic targets, and there is promising data from recent experimental and early-phase clinical studies evaluating modulators such as carnitine palmitoyltransferase 1 inhibitors, partial fatty acid oxidation inhibitors and mitochondrial-targeted antioxidants. Metabolic modulation may provide significant symptomatic and prognostic benefit for patients suffering from heart failure above and beyond guideline-directed therapy, but further clinical trials are needed.
Collapse
Affiliation(s)
- Hannah Noordali
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Brodie L Loudon
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
67
|
Tolosa L, Jiménez N, Pérez G, Castell JV, Gómez-Lechón MJ, Donato MT. Customised in vitro model to detect human metabolism-dependent idiosyncratic drug-induced liver injury. Arch Toxicol 2017; 92:383-399. [PMID: 28762043 PMCID: PMC5773651 DOI: 10.1007/s00204-017-2036-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022]
Abstract
Drug-induced liver injury (DILI) has a considerable impact on human health and is a major challenge in drug safety assessments. DILI is a frequent cause of liver injury and a leading reason for post-approval drug regulatory actions. Considerable variations in the expression levels of both cytochrome P450 (CYP) and conjugating enzymes have been described in humans, which could be responsible for increased susceptibility to DILI in some individuals. We herein explored the feasibility of the combined use of HepG2 cells co-transduced with multiple adenoviruses that encode drug-metabolising enzymes, and a high-content screening assay to evaluate metabolism-dependent drug toxicity and to identify metabolic phenotypes with increased susceptibility to DILI. To this end, HepG2 cells with different expression levels of specific drug-metabolism enzymes (CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4, GSTM1 and UGT2B7) were exposed to nine drugs with reported hepatotoxicity. A panel of pre-lethal mechanistic parameters (mitochondrial superoxide production, mitochondrial membrane potential, ROS production, intracellular calcium concentration, apoptotic nuclei) was used. Significant differences were observed according to the level of expression and/or the combination of several drug-metabolism enzymes in the cells created ad hoc according to the enzymes implicated in drug toxicity. Additionally, the main mechanisms implicated in the toxicity of the compounds were also determined showing also differences between the different types of cells employed. This screening tool allowed to mimic the variability in drug metabolism in the population and showed a highly efficient system for predicting human DILI, identifying the metabolic phenotypes associated with increased DILI risk, and indicating the mechanisms implicated in their toxicity.
Collapse
Affiliation(s)
- Laia Tolosa
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain.
| | - Nuria Jiménez
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Gabriela Pérez
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - José V Castell
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, 46010, Valencia, Spain
| | - M José Gómez-Lechón
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - M Teresa Donato
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain. .,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, 46010, Valencia, Spain.
| |
Collapse
|
68
|
Itkonen HM, Brown M, Urbanucci A, Tredwell G, Lau CH, Barfeld S, Hart C, Guldvik IJ, Takhar M, Heemers HV, Erho N, Bloch K, Davicioni E, Derua R, Waelkens E, Mohler JL, Clarke N, Swinnen JV, Keun HC, Rekvig OP, Mills IG. Lipid degradation promotes prostate cancer cell survival. Oncotarget 2017; 8:38264-38275. [PMID: 28415728 PMCID: PMC5503531 DOI: 10.18632/oncotarget.16123] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/01/2017] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer is the most common male cancer and androgen receptor (AR) is the major driver of the disease. Here we show that Enoyl-CoA delta isomerase 2 (ECI2) is a novel AR-target that promotes prostate cancer cell survival. Increased ECI2 expression predicts mortality in prostate cancer patients (p = 0.0086). ECI2 encodes for an enzyme involved in lipid metabolism, and we use multiple metabolite profiling platforms and RNA-seq to show that inhibition of ECI2 expression leads to decreased glucose utilization, accumulation of fatty acids and down-regulation of cell cycle related genes. In normal cells, decrease in fatty acid degradation is compensated by increased consumption of glucose, and here we demonstrate that prostate cancer cells are not able to respond to decreased fatty acid degradation. Instead, prostate cancer cells activate incomplete autophagy, which is followed by activation of the cell death response. Finally, we identified a clinically approved compound, perhexiline, which inhibits fatty acid degradation, and replicates the major findings for ECI2 knockdown. This work shows that prostate cancer cells require lipid degradation for survival and identifies a small molecule inhibitor with therapeutic potential.
Collapse
Affiliation(s)
- Harri M Itkonen
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Michael Brown
- Genito Urinary Cancer Research Group, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Alfonso Urbanucci
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research and Oslo University Hospital, Oslo, Norway
| | - Gregory Tredwell
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Chung Ho Lau
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Stefan Barfeld
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Claire Hart
- Genito Urinary Cancer Research Group, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Ingrid J. Guldvik
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Mandeep Takhar
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Hannelore V. Heemers
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Urology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Hematology/Medical Oncology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicholas Erho
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Katarzyna Bloch
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI Leuven Cancer Institute, KU Leuven-University of Leuven, Leuven, Belgium
| | - Elai Davicioni
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Rita Derua
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven-University of Leuven, Leuven, Belgium
| | - Etienne Waelkens
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven-University of Leuven, Leuven, Belgium
| | - James L. Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Noel Clarke
- Genito Urinary Cancer Research Group, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- PCUK/Movember Centre of Excellence for Prostate Cancer Research, CRUK Manchester Institute for Cancer Research, University of Manchester, Manchester, UK
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK
| | - Johan V. Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI Leuven Cancer Institute, KU Leuven-University of Leuven, Leuven, Belgium
| | - Hector C. Keun
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Ole P. Rekvig
- Department of Medical Biology, University of Tromso, Tromso, Norway
| | - Ian G. Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research and Oslo University Hospital, Oslo, Norway
- PCUK/Movember Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, Belfast, UK
| |
Collapse
|
69
|
Targeting Metabolic Modulation and Mitochondrial Dysfunction in the Treatment of Heart Failure. Diseases 2017; 5:diseases5020014. [PMID: 28933367 PMCID: PMC5547981 DOI: 10.3390/diseases5020014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 12/11/2022] Open
Abstract
Despite significant improvements in morbidity and mortality with current evidence-based pharmaceutical-based treatment of heart failure (HF) over the previous decades, the burden of HF remains high. An alternative approach is currently being developed, which targets myocardial energy efficiency and the dysfunction of the cardiac mitochondria. Emerging evidence suggests that the insufficient availability of ATP to the failing myocardium can be attributed to abnormalities in the myocardial utilisation of its substrates rather than an overall lack of substrate availability. Therefore, the development of potential metabolic therapeutics has commenced including trimetazidine, ranolazine and perhexiline, as well as specific mitochondrial-targeting pharmaceuticals, such as elamipretide. Large randomised controlled trials are required to confirm the role of metabolic-modulating drugs in the treatment of heart failure, but early studies have been promising in their possible efficacy for the management of heart failure in the future.
Collapse
|
70
|
Abstract
Angina pectoris is the most prevalent symptomatic manifestation of ischemic heart disease, frequently leads to a poor quality of life, and is a major cause of medical resource consumption. Since the early descriptions of nitrite and nitrate in the 19th century, there has been considerable advancement in the pharmacologic management of angina. Areas covered: Management of chronic angina is often challenging for clinicians. Despite introduction of several pharmacological agents in last few decades, a significant proportion of patients continue to experience symptoms (i.e., refractory angina) with subsequent disability. For the purpose of this review, we searched PubMed and Cochrane databases from inception to August 2016 for the most clinically relevant publications that guide current practice in angina therapy and its development. In this article, we briefly review the pathophysiology of angina and mechanism-based classification of current therapy. This is followed by evidence-based insight into the traditional and novel pharmacotherapeutic agents, highlighting their clinical usefulness. Expert opinion: Considering the wide array of available therapies with different mechanism efficacy and limiting factors, a personalized approach is essential, particularly for patients with refractory angina. Ongoing research with novel pharmacologic modalities is likely to provide new options for management of angina.
Collapse
Affiliation(s)
- Ankur Jain
- a Department of Medicine , University of Florida , Gainesville , FL , USA
| | - Islam Y Elgendy
- a Department of Medicine , University of Florida , Gainesville , FL , USA
| | - Mohammad Al-Ani
- a Department of Medicine , University of Florida , Gainesville , FL , USA
| | - Nayan Agarwal
- a Department of Medicine , University of Florida , Gainesville , FL , USA
| | - Carl J Pepine
- a Department of Medicine , University of Florida , Gainesville , FL , USA
| |
Collapse
|
71
|
Tseng CC, Noordali H, Sani M, Madhani M, Grant DM, Frenneaux MP, Zanda M, Greig IR. Development of Fluorinated Analogues of Perhexiline with Improved Pharmacokinetic Properties and Retained Efficacy. J Med Chem 2017; 60:2780-2789. [PMID: 28277663 DOI: 10.1021/acs.jmedchem.6b01592] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We designed and synthesized perhexiline analogues that have the same therapeutic profile as the parent cardiovascular drug but lacking its metabolic liability associated with CYP2D6 metabolism. Cycloalkyl perhexiline analogues 6a-j were found to be unsuitable for further development, as they retained a pharmacokinetic profile very similar to that shown by the parent compound. Multistep synthesis of perhexiline analogues incorporating fluorine atoms onto the cyclohexyl ring(s) provided a range of different fluoroperhexiline analogues. Of these, analogues 50 (4,4-gem-difluoro) and 62 (4,4,4',4'-tetrafluoro) were highly stable and showed greatly reduced susceptibility to CYP2D6-mediated metabolism. In vitro efficacy studies demonstrated that a number of derivatives retained acceptable potency against CPT-1. Having the best balance of properties, 50 was selected for further evaluation. Like perhexiline, it was shown to be selectively concentrated in the myocardium and, using the Langendorff model, to be effective in improving both cardiac contractility and relaxation when challenged with high fat buffer.
Collapse
Affiliation(s)
- Chih-Chung Tseng
- Kosterlitz Centre for Therapeutics, University of Aberdeen , Foresterhill, Aberdeen, AB25 2ZD, U.K
| | - Hannah Noordali
- Institute of Cardiovascular Sciences, University of Birmingham , Edgbaston, Birmingham, B15 2TT, U.K
| | - Monica Sani
- C.N.R.-I.C.R.M. , Via Mancinelli 7, 20131 Milan, Italy.,KemoTech s.r.l. , Parco Scientifico della Sardegna, Edificio 3, Loc. Piscinamanna, 09010 Pula, CA, Italy
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham , Edgbaston, Birmingham, B15 2TT, U.K
| | - Denis M Grant
- Department of Pharmacology and Toxicology, University of Toronto , Toronto M5S 1A8, Canada
| | - Michael P Frenneaux
- Kosterlitz Centre for Therapeutics, University of Aberdeen , Foresterhill, Aberdeen, AB25 2ZD, U.K.,Norwich Medical School, University of East Anglia , Norwich NR4 7UQ, U.K
| | - Matteo Zanda
- Kosterlitz Centre for Therapeutics, University of Aberdeen , Foresterhill, Aberdeen, AB25 2ZD, U.K.,C.N.R.-I.C.R.M. , Via Mancinelli 7, 20131 Milan, Italy
| | - Iain R Greig
- Kosterlitz Centre for Therapeutics, University of Aberdeen , Foresterhill, Aberdeen, AB25 2ZD, U.K
| |
Collapse
|
72
|
Abstract
INTRODUCTION Angina pectoris, or symptomatic myocardial ischaemia, reflects an impairment of coronary blood flow, and usually a deficiency of available myocardial energetics. Treatment options vary with the precise cause, which may vary with regards to the roles of increased myocardial oxygen demand versus reduced supply. Traditionally, organic nitrates, β-adrenoceptor antagonists, and non-dihydropyridine calcium antagonists were the only commonly used prophylactic anti-anginal agents. However, many patients failed to respond adequately to such therapy, and/or were unsuitable for their use. Areas covered: A number of 'new' agents have been shown to represent ancillary forms of prophylactic anti-anginal therapy and are particularly useful in patients who are relatively unsuitable for either percutaneous or surgical revascularisation. These include modulators of myocardial metabolic efficiency, such as perhexiline, trimetazidine and ranolazine, as well as high dose allopurinol, nicorandil and ivabradine. The advantages and disadvantages of these various agents are summarized. Expert opinion: 'Optimal' medical treatment of angina pectoris now includes use of agents primarily intended to reduce risk of infarction (e.g. statins, aspirin, ACE inhibitors). In patients whose angina persists despite the use of 'standard' anti-anginal therapy, and who are not ideal for invasive revascularization options, a number of emerging drugs offer prospects of symptomatic relief.
Collapse
Affiliation(s)
- Cher-Rin Chong
- a Cardiology and Clinical Pharmacology Departments, Basil Hetzel Institute , Queen Elizabeth Hospital, University of Adelaide , Adelaide , SA , Australia.,b Pharmacy Department , Queen Elizabeth Hospital , Woodville South , SA , Australia
| | - Gao J Ong
- a Cardiology and Clinical Pharmacology Departments, Basil Hetzel Institute , Queen Elizabeth Hospital, University of Adelaide , Adelaide , SA , Australia
| | - John D Horowitz
- a Cardiology and Clinical Pharmacology Departments, Basil Hetzel Institute , Queen Elizabeth Hospital, University of Adelaide , Adelaide , SA , Australia
| |
Collapse
|
73
|
Evidence-based selection of training compounds for use in the mechanism-based integrated prediction of drug-induced liver injury in man. Arch Toxicol 2016; 90:2979-3003. [PMID: 27659300 PMCID: PMC5104805 DOI: 10.1007/s00204-016-1845-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 08/29/2016] [Indexed: 12/16/2022]
Abstract
The current test systems employed by pharmaceutical industry are poorly predictive for drug-induced liver injury (DILI). The ‘MIP-DILI’ project addresses this situation by the development of innovative preclinical test systems which are both mechanism-based and of physiological, pharmacological and pathological relevance to DILI in humans. An iterative, tiered approach with respect to test compounds, test systems, bioanalysis and systems analysis is adopted to evaluate existing models and develop new models that can provide validated test systems with respect to the prediction of specific forms of DILI and further elucidation of mechanisms. An essential component of this effort is the choice of compound training set that will be used to inform refinement and/or development of new model systems that allow prediction based on knowledge of mechanisms, in a tiered fashion. In this review, we focus on the selection of MIP-DILI training compounds for mechanism-based evaluation of non-clinical prediction of DILI. The selected compounds address both hepatocellular and cholestatic DILI patterns in man, covering a broad range of pharmacologies and chemistries, and taking into account available data on potential DILI mechanisms (e.g. mitochondrial injury, reactive metabolites, biliary transport inhibition, and immune responses). Known mechanisms by which these compounds are believed to cause liver injury have been described, where many if not all drugs in this review appear to exhibit multiple toxicological mechanisms. Thus, the training compounds selection offered a valuable tool to profile DILI mechanisms and to interrogate existing and novel in vitro systems for the prediction of human DILI.
Collapse
|
74
|
The standardized functional observational battery: Its intrinsic value remains in the instrument of measure: The rat. J Pharmacol Toxicol Methods 2016; 82:90-108. [PMID: 27534836 DOI: 10.1016/j.vascn.2016.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/10/2016] [Accepted: 08/01/2016] [Indexed: 12/13/2022]
Abstract
The International Conference on Harmonisation's (ICH) Tripartite Guideline on Safety Pharmacology Studies for Human Pharmaceuticals has adopted the requirement that each new test substance must be tested for effects on the central nervous system prior to "first dose in man". This assessment is required to measure, at a minimum, the effects of the substance on general motor activity, behavioral changes, coordination, sensory/motor reflex responses, and body temperatures. To achieve this goal, ICH S7A recommends a neurobehavioral assessment (usually a functional observational battery (FOB) or modified Irwin test), which is generally undertaken in the rat. There seems to be a growing lack of consensus on the value of the FOB to determine CNS safety. This review highlights the importance of the time, effort and cost of training technicians to familiarize with their instrument of measure, so that each observer is better able to identify and document very subtle changes in behavior that will serve to increase the reliability and validity of these assays with respect to CNS safety assessments.
Collapse
|
75
|
Guidi A, Lalli C, Perlas E, Bolasco G, Nibbio M, Monteagudo E, Bresciani A, Ruberti G. Discovery and Characterization of Novel Anti-schistosomal Properties of the Anti-anginal Drug, Perhexiline and Its Impact on Schistosoma mansoni Male and Female Reproductive Systems. PLoS Negl Trop Dis 2016; 10:e0004928. [PMID: 27518281 PMCID: PMC4982595 DOI: 10.1371/journal.pntd.0004928] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 07/26/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Schistosomiasis, one of the world's greatest human neglected tropical diseases, is caused by parasitic trematodes of the genus Schistosoma. A unique feature of schistosome biology is that the induction of sexual maturation as well as the maintenance of the differentiation status of female reproductive organs and egg production, necessary for both disease transmission and pathogenesis, are strictly dependent on the male. The treatment and most control initiatives of schistosomiasis rely today on the long-term application of a single drug, praziquantel (PZQ), mostly by campaigns of mass drug administration. PZQ, while very active on adult parasites, has much lower activity against juvenile worms. Monotherapy also favors the selection of drug resistance and, therefore, new drugs are urgently needed. METHODS AND FINDINGS Following the screening of a small compound library with an ATP-based luminescent assay on Schistosoma mansoni schistosomula, we here report the identification and characterization of novel antischistosomal properties of the anti-anginal drug perhexiline maleate (PHX). By phenotypic worm survival assays and confocal microscopy studies we show that PHX, in vitro, has a marked lethal effect on all S. mansoni parasite life stages (newly transformed schistosomula, juvenile and adult worms) of the definitive host. We further demonstrate that sub-lethal doses of PHX significantly impair egg production and lipid depletion within the vitellarium of adult female worms. Moreover, we highlighted tegumental damage in adult male worms and remarkable reproductive system alterations in both female and male adult parasites. The in vivo study in S. mansoni-patent mice showed a notable variability of worm burdens in the individual experiments, with an overall minimal schistosomicidal effect upon PHX treatment. The short PHX half-life in mice, together with its very high rodent plasma proteins binding could be the cause of the modest efficacy of PHX in the schistosomiasis murine model. CONCLUSIONS/SIGNIFICANCE Overall, our data indicate that PHX could represent a promising starting point for novel schistosomicidal drug discovery programmes.
Collapse
Affiliation(s)
- Alessandra Guidi
- National Research Council, Institute of Cell Biology and Neurobiology, Campus A. Buzzati-Traverso Monterotondo, Roma, Italy
| | - Cristiana Lalli
- National Research Council, Institute of Cell Biology and Neurobiology, Campus A. Buzzati-Traverso Monterotondo, Roma, Italy
| | - Emerald Perlas
- European Molecular Biology Laboratory (EMBL), Mouse Biology Unit, Monterotondo, Italy
| | - Giulia Bolasco
- European Molecular Biology Laboratory (EMBL), Mouse Biology Unit, Monterotondo, Italy
| | - Martina Nibbio
- IRBM Science Park, Department of Preclinical Research, Pomezia, Italy
| | - Edith Monteagudo
- IRBM Science Park, Department of Preclinical Research, Pomezia, Italy
| | | | - Giovina Ruberti
- National Research Council, Institute of Cell Biology and Neurobiology, Campus A. Buzzati-Traverso Monterotondo, Roma, Italy
- * E-mail:
| |
Collapse
|
76
|
Oorts M, Baze A, Bachellier P, Heyd B, Zacharias T, Annaert P, Richert L. Drug-induced cholestasis risk assessment in sandwich-cultured human hepatocytes. Toxicol In Vitro 2016; 34:179-186. [DOI: 10.1016/j.tiv.2016.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/09/2016] [Accepted: 03/11/2016] [Indexed: 12/19/2022]
|
77
|
Sherrid MV. Drug Therapy for Hypertrophic Cardiomypathy: Physiology and Practice. Curr Cardiol Rev 2016; 12:52-65. [PMID: 26818487 PMCID: PMC4807719 DOI: 10.2174/1573403x1201160126125403] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 12/19/2022] Open
Abstract
HCM is the most common inherited heart condition occurring in 1:500 individuals in the general population. Left ventricular outflow obstruction at rest or after provocation occurs in 2/3 of HCM patients and is a frequent cause of limiting symptoms. Pharmacologic therapy is the first-line treatment for obstruction, and should be aggressively pursued before application of invasive therapy. Beta-blockade is given first, and up-titrated to decrease resting heart rate to between 50 and 60 beats per minute. However, beta-blockade is not expected to decrease resting gradients; its effect rests on decreasing the rise in gradient that accompanies exercise. For patients who fail beta-blockade the addition of oral disopyramide in adequate dose often will decrease resting gradients and offer meaningful relief of symptoms. Disopyramide vagolytic side effects, if they occur, can be greatly mitigated by simultaneous administration of oral pyridostigmine. This combination allows adequate dosing of disopyramide to achieve therapeutic goals. Verapamil utility in obstructive HCM with high resting gradients is limited by its vasodilating effects that can, infrequently, worsen gradient and symptoms. As such, we tend to avoid it in patients with high gradients and limiting heart failure symptoms. In a head-to-head comparison of intravenous drug administration in individual obstructive HCM patients the relative efficacy for lowering gradient was disopyramide > beta-blockade > verapamil. Severe symptoms in non-obstructive HCM are caused by fibrosis or severe myocyte disarray, and often by very small LV chamber size. Severe symptoms caused by these anatomic and histologic abnormalities, in the absence of obstruction, are less amenable to current pharmacotherapy. New pharmacotherapeutic approaches to HCM are on the horizon, that are to be evaluated in formal therapeutic trials.
Collapse
Affiliation(s)
- Mark V Sherrid
- New York University Langone Medical Center, 530 First Avenue, NYC, NY 10016, USA.
| |
Collapse
|
78
|
George CH, Mitchell AN, Preece R, Bannister ML, Yousef Z. Pleiotropic mechanisms of action of perhexiline in heart failure. Expert Opin Ther Pat 2016; 26:1049-59. [PMID: 27455171 DOI: 10.1080/13543776.2016.1211111] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The re-purposing of the anti-anginal drug perhexiline (PHX) has resulted in symptomatic improvements in heart failure (HF) patients. The inhibition of carnitine palmitoyltransferase-1 (CPT-1) has been proposed as the primary mechanism underlying the therapeutic benefit of PHX. This hypothesis is contentious. AREAS COVERED We reviewed the primary literature and patent landscape of PHX from its initial development in the 1960s through to its emergence as a drug beneficial for HF. We focused on its physico-chemistry, molecular targets, tissue accumulation and clinical dosing. EXPERT OPINION Dogma that the beneficial effects of PHX are due primarily to potent myocardial CPT-1 inhibition is not supported by the literature and all available evidence point to it being extremely unlikely that the major effects of PHX occur via this mechanism. In vivo PHX is much more likely to be an inhibitor of surface membrane ion channels and also to have effects on other components of cellular metabolism and reactive oxygen species (ROS) generation across the cardiovascular system. However, the possibility that minor effects of PHX on CPT-1 underpin disproportionately large effects on myocardial function cannot be entirely excluded, especially given the massive accumulation of the drug in heart tissue.
Collapse
Affiliation(s)
- Christopher H George
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Alice N Mitchell
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Ryan Preece
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Mark L Bannister
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| | - Zaheer Yousef
- a Wales Heart Research Institute, School of Medicine , Cardiff University , Cardiff , UK
| |
Collapse
|
79
|
Aberrant Lipid Metabolism Promotes Prostate Cancer: Role in Cell Survival under Hypoxia and Extracellular Vesicles Biogenesis. Int J Mol Sci 2016; 17:ijms17071061. [PMID: 27384557 PMCID: PMC4964437 DOI: 10.3390/ijms17071061] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer (PCa) is the leading malignancy among men in United States. Recent studies have focused on the identification of novel metabolic characteristics of PCa, aimed at devising better preventive and therapeutic approaches. PCa cells have revealed unique metabolic features such as higher expression of several enzymes associated with de novo lipogenesis, fatty acid up-take and β-oxidation. This aberrant lipid metabolism has been reported to be important for PCa growth, hormone-refractory progression and treatment resistance. Furthermore, PCa cells effectively use lipid metabolism under adverse environmental conditions for their survival advantage. Specifically, hypoxic cancer cells accumulate higher amount of lipids through a combination of metabolic alterations including high glutamine and fatty acid uptake, as well as decreased fatty acid oxidation. These stored lipids serve to protect cancer cells from oxidative and endoplasmic reticulum stress, and play important roles in fueling cancer cell proliferation following re-oxygenation. Lastly, cellular lipids have also been implicated in extracellular vesicle biogenesis, which play a vital role in intercellular communication. Overall, the new understanding of lipid metabolism in recent years has offered several novel targets to better target and manage clinical PCa.
Collapse
|
80
|
Giannopoulos AA, Giannoglou GD, Chatzizisis YS. Pharmacological approaches of refractory angina. Pharmacol Ther 2016; 163:118-31. [DOI: 10.1016/j.pharmthera.2016.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
81
|
Fatty acid oxidation and carnitine palmitoyltransferase I: emerging therapeutic targets in cancer. Cell Death Dis 2016; 7:e2226. [PMID: 27195673 PMCID: PMC4917665 DOI: 10.1038/cddis.2016.132] [Citation(s) in RCA: 361] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 04/07/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022]
Abstract
Tumor cells exhibit unique metabolic adaptations that are increasingly viewed as potential targets for novel and specific cancer therapies. Among these targets, the carnitine palmitoyltransferase system is responsible for delivering the long-chain fatty acid (FA) from cytoplasm into mitochondria for oxidation, where carnitine palmitoyltransferase I (CPTI) catalyzes the rate-limiting step of fatty acid oxidation (FAO). With increasing understanding of the crucial role had by fatty acid oxidation in cancer, CPTI has received renewed attention as a pivotal mediator in cancer metabolic mechanism. CPTI activates FAO and fuels cancer growth via ATP and NADPH production, constituting an essential part of cancer metabolism adaptation. Moreover, CPTI also functionally intertwines with other key pathways and factors to regulate gene expression and apoptosis of cancer cell. Here, we summarize recent findings and update the current understanding of FAO and CPTI in cancer and provide theoretical basis for this enzyme as an emerging potential molecular target in cancer therapeutic intervention.
Collapse
|
82
|
Loudon BL, Noordali H, Gollop ND, Frenneaux MP, Madhani M. Present and future pharmacotherapeutic agents in heart failure: an evolving paradigm. Br J Pharmacol 2016; 173:1911-24. [PMID: 26993743 PMCID: PMC4882493 DOI: 10.1111/bph.13480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/28/2016] [Accepted: 02/26/2016] [Indexed: 02/06/2023] Open
Abstract
Many conditions culminate in heart failure (HF), a multi‐organ systemic syndrome with an intrinsically poor prognosis. Pharmacotherapeutic agents that correct neurohormonal dysregulation and haemodynamic instability have occupied the forefront of developments within the treatment of HF in the past. Indeed, multiple trials aimed to validate these agents in the 1980s and early 1990s, resulting in a large and robust evidence‐base supporting their use clinically. An established treatment paradigm now exists for the treatment of HF with reduced ejection fraction (HFrEF), but there have been very few notable developments in recent years. HF remains a significant health concern with an increasing incidence as the population ages. We may indeed be entering the surgical era for HF treatment, but these therapies remain expensive and inaccessible to many. Newer pharmacotherapeutic agents are slowly emerging, many targeting alternative therapeutic pathways, but with mixed results. Metabolic modulation and manipulation of the nitrate/nitrite/nitric oxide pathway have shown promise and could provide the answers to fill the therapeutic gap between medical interventions and surgery, but further definitive trials are warranted. We review the significant evidence base behind the current medical treatments for HFrEF, the physiology of metabolic impairment in HF, and discuss two promising novel agents, perhexiline and nitrite.
Collapse
Affiliation(s)
- Brodie L Loudon
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Hannah Noordali
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nicholas D Gollop
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Michael P Frenneaux
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
83
|
Tolosa L, Gómez-Lechón MJ, Jiménez N, Hervás D, Jover R, Donato MT. Advantageous use of HepaRG cells for the screening and mechanistic study of drug-induced steatosis. Toxicol Appl Pharmacol 2016; 302:1-9. [PMID: 27089845 DOI: 10.1016/j.taap.2016.04.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/11/2022]
Abstract
Only a few in vitro assays have been proposed to evaluate the steatotic potential of new drugs. The present study examines the utility of HepaRG cells as a cell-based assay system for screening drug-induced liver steatosis. A high-content screening assay was run to evaluate multiple toxicity-related cell parameters in HepaRG cells exposed to 28 compounds, including drugs reported to cause steatosis through different mechanisms and non-steatotic compounds. Lipid content was the most sensitive parameter for all the steatotic drugs, whereas no effects on lipid levels were produced by non-steatotic compounds. Apart from fat accumulation, increased ROS production and altered mitochondrial membrane potential were also found in the cells exposed to steatotic drugs, which indicates that all these cellular events contributed to drug-induced hepatotoxicity. These findings are of clinical relevance as most effects were observed at drug concentrations under 100-fold of the therapeutic peak plasmatic concentration. HepaRG cells showed increased lipid overaccumulation vs. HepG2 cells, which suggests greater sensitivity to drug-induced steatosis. An altered expression profile of transcription factors and the genes that code key proteins in lipid metabolism was also found in the cells exposed to drugs capable of inducing liver steatosis. Our results generally indicate the value of HepaRG cells for assessing the risk of liver damage associated with steatogenic compounds and for investigating the molecular mechanisms involved in drug-induced steatosis.
Collapse
Affiliation(s)
- Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain
| | - M José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain; CIBERehd, FIS, Barcelona 08036, Spain
| | - Nuria Jiménez
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain
| | - David Hervás
- Biostatistics Unit, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain
| | - Ramiro Jover
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain; CIBERehd, FIS, Barcelona 08036, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia 46010, Spain
| | - M Teresa Donato
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain; CIBERehd, FIS, Barcelona 08036, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia 46010, Spain.
| |
Collapse
|
84
|
Gehmlich K, Dodd MS, Allwood JW, Kelly M, Bellahcene M, Lad HV, Stockenhuber A, Hooper C, Ashrafian H, Redwood CS, Carrier L, Dunn WB. Changes in the cardiac metabolome caused by perhexiline treatment in a mouse model of hypertrophic cardiomyopathy. MOLECULAR BIOSYSTEMS 2016; 11:564-73. [PMID: 25437646 DOI: 10.1039/c4mb00594e] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Energy depletion has been highlighted as an important contributor to the pathology of hypertrophic cardiomyopathy (HCM), a common inherited cardiac disease. Pharmacological reversal of energy depletion appears an attractive approach and the use of perhexiline has been proposed as it is thought to shift myocardial metabolism from fatty acid to glucose utilisation, increasing ATP production and myocardial efficiency. We used the Mybpc3-targeted knock-in mouse model of HCM to investigate changes in the cardiac metabolome following perhexiline treatment. Echocardiography indicated that perhexiline induced partial improvement of some, but not all hypertrophic parameters after six weeks. Non-targeted metabolomics, applying ultra-high performance liquid chromatography-mass spectrometry, described a phenotypic modification of the cardiac metabolome with 272 unique metabolites showing a statistically significant change (p < 0.05). Changes in fatty acids and acyl carnitines indicate altered fatty acid transport into mitochondria, implying reduction in fatty acid beta-oxidation. Increased glucose utilisation is indirectly implied through changes in the glycolytic, glycerol, pentose phosphate, tricarboxylic acid and pantothenate pathways. Depleted reduced glutathione and increased production of NADPH suggest reduction in oxidative stress. These data delineate the metabolic changes occurring during improvement of the HCM phenotype and indicate the requirements for further targeted interventions.
Collapse
Affiliation(s)
- Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Elleraas J, Ewanicki J, Johnson TW, Sach NW, Collins MR, Richardson PF. Conformational Studies and Atropisomerism Kinetics of the ALK Clinical Candidate Lorlatinib (PF-06463922) and Desmethyl Congeners. Angew Chem Int Ed Engl 2016; 55:3590-5. [DOI: 10.1002/anie.201509240] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 11/27/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Jeff Elleraas
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Jason Ewanicki
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Ted W. Johnson
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Neal W. Sach
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Michael R. Collins
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Paul F. Richardson
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| |
Collapse
|
86
|
Elleraas J, Ewanicki J, Johnson TW, Sach NW, Collins MR, Richardson PF. Conformational Studies and Atropisomerism Kinetics of the ALK Clinical Candidate Lorlatinib (PF-06463922) and Desmethyl Congeners. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201509240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jeff Elleraas
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Jason Ewanicki
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Ted W. Johnson
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Neal W. Sach
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Michael R. Collins
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| | - Paul F. Richardson
- Oncology Medicinal Chemistry; Pfizer, La Jolla; 10770 Science Center Drive San Diego CA 92121 USA
| |
Collapse
|
87
|
|
88
|
Esposito S, Bracacel E, Nibbio M, Speziale R, Orsatti L, Veneziano M, Monteagudo E, Bonelli F. Use of 'dilute-and-shoot' liquid chromatography-high resolution mass spectrometry in preclinical research: application to a DMPK study of perhexiline in mouse plasma. J Pharm Biomed Anal 2015; 118:70-80. [PMID: 26517851 DOI: 10.1016/j.jpba.2015.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/02/2015] [Accepted: 10/04/2015] [Indexed: 01/20/2023]
Abstract
This work describes a simple, sensitive and rapid liquid chromatography-high resolution mass spectrometry method for the quantitation of perhexiline and the simultaneous detection of perhexiline metabolites in C57bl/6 mice plasma. Only 5 μL of plasma was used for analysis. Pretreatment was limited to a 100-fold dilution ('dilute-and-shoot'). The analyte was detected by high resolution mass spectrometry (Orbitrap™ technology). Three scan events were performed over the entire chromatogram. Targeted single ion monitoring with data dependent acquisition was employed for perhexiline quantitation and confirmation, while full scan was used to perform untargeted detection of perhexiline phase I and phase II circulating metabolites. The calibration curve was linear (r(2)=0.990) ranging from 0.305 ng/mL (LLOQ) to 10000 ng/mL. Matrix effect was limited to 6.1%. The method was applied to a pharmacokinetic study of perhexiline in mouse plasma and the results obtained were compared to a standard sample preparation method based on protein precipitation and liquid chromatography-tandem mass spectrometry (MRM mode) detection. The new approach provided comparable results in terms of pharmacokinetics parameters estimate with a high sensitivity, additional information on perhexiline circulating metabolites and a low consumption of biological sample. The combination of the 'dilute-and-shoot' approach together with HRMS targeted and untargeted detection represents a suitable alternative to classic bioanalytical approaches in preclinical research.
Collapse
Affiliation(s)
- Simone Esposito
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy.
| | - Elena Bracacel
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| | - Martina Nibbio
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| | - Roberto Speziale
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| | - Laura Orsatti
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| | - Maria Veneziano
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| | - Edith Monteagudo
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| | - Fabio Bonelli
- IRBM Science Park, Via Pontina km 30,600, 00040 Pomezia, Roma, Italy
| |
Collapse
|
89
|
Diener ME, Metrano AJ, Kusano S, Miller SJ. Enantioselective synthesis of 3-arylquinazolin-4(3H)-ones via peptide-catalyzed atroposelective bromination. J Am Chem Soc 2015; 137:12369-77. [PMID: 26343278 PMCID: PMC5134330 DOI: 10.1021/jacs.5b07726] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We report the development of a tertiary amine-containing β-turn peptide that catalyzes the atroposelective bromination of pharmaceutically relevant 3-arylquinazolin-4(3H)-ones (quinazolinones) with high levels of enantioinduction over a broad substrate scope. The structure of the free catalyst and the peptide-substrate complex were explored using X-ray crystallography and 2D-NOESY experiments. Quinazolinone rotational barriers about the chiral anilide axis were also studied using density functional theory calculations and are discussed in light of the high enantioselectivities observed. Mechanistic studies also suggest that the initial bromination event is stereodetermining, and the major monobromide intermediate is an atropisomerically stable, mono-ortho-substituted isomer. The observation of stereoisomerically stable monobromides stimulated the conversion of the tribromide products to other atropisomerically defined products of interest. For example, (1) a dehalogenation Suzuki-Miyaura cross-coupling sequence delivers ortho-arylated derivatives, and (2) a regioselective Buchwald-Hartwig amination procedure installs para-amine functionality. Stereochemical information was retained during these subsequent transformations.
Collapse
Affiliation(s)
| | | | | | - Scott J. Miller
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, United States
| |
Collapse
|
90
|
Lee EA, Angka L, Rota SG, Hanlon T, Mitchell A, Hurren R, Wang XM, Gronda M, Boyaci E, Bojko B, Minden M, Sriskanthadevan S, Datti A, Wrana JL, Edginton A, Pawliszyn J, Joseph JW, Quadrilatero J, Schimmer AD, Spagnuolo PA. Targeting Mitochondria with Avocatin B Induces Selective Leukemia Cell Death. Cancer Res 2015; 75:2478-88. [PMID: 26077472 DOI: 10.1158/0008-5472.can-14-2676] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment regimens for acute myeloid leukemia (AML) continue to offer weak clinical outcomes. Through a high-throughput cell-based screen, we identified avocatin B, a lipid derived from avocado fruit, as a novel compound with cytotoxic activity in AML. Avocatin B reduced human primary AML cell viability without effect on normal peripheral blood stem cells. Functional stem cell assays demonstrated selectivity toward AML progenitor and stem cells without effects on normal hematopoietic stem cells. Mechanistic investigations indicated that cytotoxicity relied on mitochondrial localization, as cells lacking functional mitochondria or CPT1, the enzyme that facilitates mitochondria lipid transport, were insensitive to avocatin B. Furthermore, avocatin B inhibited fatty acid oxidation and decreased NADPH levels, resulting in ROS-dependent leukemia cell death characterized by the release of mitochondrial proteins, apoptosis-inducing factor, and cytochrome c. This study reveals a novel strategy for selective leukemia cell eradication based on a specific difference in mitochondrial function.
Collapse
Affiliation(s)
- Eric A Lee
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Leonard Angka
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Sarah-Grace Rota
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Thomas Hanlon
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Andrew Mitchell
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Rose Hurren
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Xiao Ming Wang
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Ezel Boyaci
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Barbara Bojko
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Mark Minden
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | | | - Alessandro Datti
- SMART Laboratory for High-Throughput Screening Programs, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada. Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Jeffery L Wrana
- SMART Laboratory for High-Throughput Screening Programs, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Jamie W Joseph
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Paul A Spagnuolo
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada.
| |
Collapse
|
91
|
Guo Y, Fan Y, Zhang J, Lomberk GA, Zhou Z, Sun L, Mathison AJ, Garcia-Barrio MT, Zhang J, Zeng L, Li L, Pennathur S, Willer CJ, Rader DJ, Urrutia R, Chen YE. Perhexiline activates KLF14 and reduces atherosclerosis by modulating ApoA-I production. J Clin Invest 2015; 125:3819-30. [PMID: 26368306 DOI: 10.1172/jci79048] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 08/07/2015] [Indexed: 12/20/2022] Open
Abstract
Recent genome-wide association studies have revealed that variations near the gene locus encoding the transcription factor Krüppel-like factor 14 (KLF14) are strongly associated with HDL cholesterol (HDL-C) levels, metabolic syndrome, and coronary heart disease. However, the precise mechanisms by which KLF14 regulates lipid metabolism and affects atherosclerosis remain largely unexplored. Here, we report that KLF14 is dysregulated in the liver of 2 dyslipidemia mouse models. We evaluated the effects of both KLF14 overexpression and genetic inactivation and determined that KLF14 regulates plasma HDL-C levels and cholesterol efflux capacity by modulating hepatic ApoA-I production. Hepatic-specific Klf14 deletion in mice resulted in decreased circulating HDL-C levels. In an attempt to pharmacologically target KLF14 as an experimental therapeutic approach, we identified perhexiline, an approved therapeutic small molecule presently in clinical use to treat angina and heart failure, as a KLF14 activator. Indeed, in WT mice, treatment with perhexiline increased HDL-C levels and cholesterol efflux capacity via KLF14-mediated upregulation of ApoA-I expression. Moreover, perhexiline administration reduced atherosclerotic lesion development in apolipoprotein E-deficient mice. Together, these results provide comprehensive insight into the KLF14-dependent regulation of HDL-C and subsequent atherosclerosis and indicate that interventions that target the KLF14 pathway should be further explored for the treatment of atherosclerosis.
Collapse
|
92
|
Singh S, Beadle R, Cameron D, Rudd A, Bruce M, Jagpal B, Schwarz K, Brindley G, Mckiddie F, Nightingale P, Lang C, Dawson D, Frenneaux M. Randomized double-blind placebo-controlled trial of perhexiline in heart failure with preserved ejection fraction syndrome. Future Cardiol 2015; 10:693-8. [PMID: 25495811 DOI: 10.2217/fca.14.62] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recently heart failure with preserved ejection fraction (HFpEF) has emerged as a huge epidemic. Increasing evidence shows the role of energy deficiency in the pathophysiology of HFpEF. In the current study, we hypothesize that the use of metabolic modulator perhexiline would correct myocardial energy deficiency and improve exercise capacity and diastolic abnormalities in patients with this syndrome.
Collapse
Affiliation(s)
- Satnam Singh
- School of Medicine & Dentistry, University of Aberdeen, Polwarth Building, Aberdeen, AB25 2ZD, Scotland, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Rosano GM, Vitale C, Spoletini I. Metabolic approach to heart failure: The role of metabolic modulators. Egypt Heart J 2015. [DOI: 10.1016/j.ehj.2015.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
94
|
Abstract
Stable angina pectoris is characterised by typical exertional chest pain that is relieved by rest or nitrates. Risk stratification of patients is important to define prognosis, to guide medical management and to select patients suitable for revascularisation. Medical treatment aims to relieve angina and prevent cardiovascular events. Beta blockers and calcium channel antagonists are first-line options for treatment. Short-acting nitrates can be used for symptom relief. Low-dose aspirin and statins are prescribed to prevent cardiovascular events.
Collapse
Affiliation(s)
- Yong Wee
- Heart Lung Institute, Prince Charles Hospital, Brisbane
| | - Kylie Burns
- Heart Lung Institute, Prince Charles Hospital, Brisbane
| | - Nicholas Bett
- Heart Lung Institute, Prince Charles Hospital, Brisbane
| |
Collapse
|
95
|
Singh S, Schwarz K, Horowitz J, Frenneaux M. Cardiac energetic impairment in heart disease and the potential role of metabolic modulators: a review for clinicians. ACTA ACUST UNITED AC 2015; 7:720-8. [PMID: 25518045 DOI: 10.1161/circgenetics.114.000221] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac energetic impairment is a frequent finding in patients with both inherited and acquired diseases of heart muscle. In this review the mechanisms of energy generation in the healthy heart and their disturbances in heart muscle diseases are described. Therapeutic agents targeted at correcting cardiac energetic impairment are discussed.
Collapse
Affiliation(s)
- Satnam Singh
- From the Department of Cardiovascular Medicine, University of Aberdeen, Aberdeen, United Kingdom (S.S., K.S., M.F.); and Cardiology Unit, The Queen Elizabeth Hospital, Adelaide, Australia (J.H.)
| | - Konstantin Schwarz
- From the Department of Cardiovascular Medicine, University of Aberdeen, Aberdeen, United Kingdom (S.S., K.S., M.F.); and Cardiology Unit, The Queen Elizabeth Hospital, Adelaide, Australia (J.H.)
| | - John Horowitz
- From the Department of Cardiovascular Medicine, University of Aberdeen, Aberdeen, United Kingdom (S.S., K.S., M.F.); and Cardiology Unit, The Queen Elizabeth Hospital, Adelaide, Australia (J.H.)
| | - Michael Frenneaux
- From the Department of Cardiovascular Medicine, University of Aberdeen, Aberdeen, United Kingdom (S.S., K.S., M.F.); and Cardiology Unit, The Queen Elizabeth Hospital, Adelaide, Australia (J.H.).
| |
Collapse
|
96
|
Rodríguez-Enríquez S, Hernández-Esquivel L, Marín-Hernández A, El Hafidi M, Gallardo-Pérez JC, Hernández-Reséndiz I, Rodríguez-Zavala JS, Pacheco-Velázquez SC, Moreno-Sánchez R. Mitochondrial free fatty acid β-oxidation supports oxidative phosphorylation and proliferation in cancer cells. Int J Biochem Cell Biol 2015; 65:209-21. [PMID: 26073129 DOI: 10.1016/j.biocel.2015.06.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/29/2015] [Accepted: 06/08/2015] [Indexed: 12/26/2022]
Abstract
Oxidative phosphorylation (OxPhos) is functional and sustains tumor proliferation in several cancer cell types. To establish whether mitochondrial β-oxidation of free fatty acids (FFAs) contributes to cancer OxPhos functioning, its protein contents and enzyme activities, as well as respiratory rates and electrical membrane potential (ΔΨm) driven by FFA oxidation were assessed in rat AS-30D hepatoma and liver (RLM) mitochondria. Higher protein contents (1.4-3 times) of β-oxidation (CPT1, SCAD) as well as proteins and enzyme activities (1.7-13-times) of Krebs cycle (KC: ICD, 2OGDH, PDH, ME, GA), and respiratory chain (RC: COX) were determined in hepatoma mitochondria vs. RLM. Although increased cholesterol content (9-times vs. RLM) was determined in the hepatoma mitochondrial membranes, FFAs and other NAD-linked substrates were oxidized faster (1.6-6.6 times) by hepatoma mitochondria than RLM, maintaining similar ΔΨm values. The contents of β-oxidation, KC and RC enzymes were also assessed in cells. The mitochondrial enzyme levels in human cervix cancer HeLa and AS-30D cells were higher than those observed in rat hepatocytes whereas in human breast cancer biopsies, CPT1 and SCAD contents were lower than in human breast normal tissue. The presence of CPT1 and SCAD in AS-30D mitochondria and HeLa cells correlated with an active FFA utilization in HeLa cells. Furthermore, the β-oxidation inhibitor perhexiline blocked FFA utilization, OxPhos and proliferation in HeLa and other cancer cells. In conclusion, functional mitochondria supported by FFA β-oxidation are essential for the accelerated cancer cell proliferation and hence anti-β-oxidation therapeutics appears as an alternative promising approach to deter malignant tumor growth.
Collapse
Affiliation(s)
- Sara Rodríguez-Enríquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico; Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Ciudad de Mexico, D.F., Mexico.
| | - Luz Hernández-Esquivel
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| | - Alvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| | - Mohammed El Hafidi
- Departamento de Medicina Cardiovascular, Instituto Nacional de Cardiología, Ciudad de México, D.F., Mexico
| | - Juan Carlos Gallardo-Pérez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| | - Ileana Hernández-Reséndiz
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| | - José S Rodríguez-Zavala
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| | - Silvia C Pacheco-Velázquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| | - Rafael Moreno-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Sección 16, Tlalpan, México D.F. 14080, Mexico
| |
Collapse
|
97
|
Schwarz EM, Hu Y, Antoshechkin I, Miller MM, Sternberg PW, Aroian RV. The genome and transcriptome of the zoonotic hookworm Ancylostoma ceylanicum identify infection-specific gene families. Nat Genet 2015; 47:416-22. [PMID: 25730766 PMCID: PMC4617383 DOI: 10.1038/ng.3237] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 02/05/2015] [Indexed: 12/19/2022]
Abstract
Hookworms infect over 400 million people, stunting and impoverishing them. Sequencing hookworm genomes and finding which genes they express during infection should help in devising new drugs or vaccines against hookworms. Unlike other hookworms, Ancylostoma ceylanicum infects both humans and other mammals, providing a laboratory model for hookworm disease. We determined an A. ceylanicum genome sequence of 313 Mb, with transcriptomic data throughout infection showing expression of 30,738 genes. Approximately 900 genes were upregulated during early infection in vivo, including ASPRs, a cryptic subfamily of activation-associated secreted proteins (ASPs). Genes downregulated during early infection included ion channels and G protein-coupled receptors; this downregulation was observed in both parasitic and free-living nematodes. Later, at the onset of heavy blood feeding, C-lectin genes were upregulated along with genes for secreted clade V proteins (SCVPs), encoding a previously undescribed protein family. These findings provide new drug and vaccine targets and should help elucidate hookworm pathogenesis.
Collapse
Affiliation(s)
- Erich M Schwarz
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Yan Hu
- 1] Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA. [2] Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, USA
| | - Igor Antoshechkin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Melanie M Miller
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, USA
| | - Paul W Sternberg
- 1] Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA. [2] Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, USA
| | - Raffi V Aroian
- 1] Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA. [2] Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
98
|
Tham YK, Bernardo BC, Ooi JYY, Weeks KL, McMullen JR. Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets. Arch Toxicol 2015; 89:1401-38. [DOI: 10.1007/s00204-015-1477-x] [Citation(s) in RCA: 371] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 12/18/2022]
|
99
|
Perhexiline: lessons for heart failure therapeutics. JACC-HEART FAILURE 2015; 3:212-3. [PMID: 25650368 DOI: 10.1016/j.jchf.2014.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/15/2014] [Indexed: 11/21/2022]
|
100
|
O'Sullivan D, Pearce EL. Targeting T cell metabolism for therapy. Trends Immunol 2015; 36:71-80. [PMID: 25601541 DOI: 10.1016/j.it.2014.12.004] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 12/14/2014] [Accepted: 12/15/2014] [Indexed: 12/13/2022]
Abstract
In the past several years a wealth of evidence has emerged illustrating how metabolism supports many aspects of T cell biology, as well as how metabolic changes drive T cell differentiation and fate. We outline developing principles in the regulation of T cell metabolism, and discuss how these processes are affected in settings of inflammation and cancer. In this context we discuss how metabolic pathways might be manipulated for the treatment of human disease, including how metabolism may be targeted to prevent T cell dysfunction in inhospitable microenvironments, to generate more effective adoptive cellular immunotherapies in cancer, and to direct T cell differentiation and function towards non-pathogenic phenotypes in settings of autoimmunity.
Collapse
Affiliation(s)
- David O'Sullivan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Erika L Pearce
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|