1
|
Liu X, Lai YC, Cui D, Kung SC, Park M, Zoltan L, Larson PE, Wang ZJ. Initial Experience of Metabolic Imaging with Hyperpolarized [1- 13C]pyruvate MRI in Kidney Transplant Patients. ARXIV 2024:arXiv:2409.06664v1. [PMID: 39314508 PMCID: PMC11419194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
BACKGROUND Kidney transplant is the treatment of choice for patients with end-stage renal disease. Early detection of allograft injury is important to delay or prevent irreversible damage. PURPOSE To investigate the feasibility of hyperpolarized (HP) [1-13C]pyruvate MRI for assessing kidney allograft metabolism. STUDY TYPE Prospective. SUBJECTS 6 participants (mean age, 45.2 ± 12.4 years, 2 females) scheduled for kidney allograft biopsy and 5 patients (mean age, 59.6 ± 10.4 years, 2 females) with renal cell carcinoma (RCC). FIELD STRENGTH/SEQUENCE 3 Tesla, T2-weighted fast spin echo, multi-echo gradient echo, single shot diffusion-weighted echo-planar imaging, and time-resolved HP 13C metabolite-selective imaging. ASSESSMENT Five of the six kidney allograft participants underwent biopsy after MRI. Estimated glomerular filtration rate (eGFR) and urine protein-to-creatine ratio (uPCR) were collected within 4 weeks of MRI. Kidney metabolism was quantified from HP [1-13C]pyruvate MRI using the lactate-to-pyruvate ratio in allograft kidneys and non-tumor bearing kidneys from RCC patients. STATISTICAL TESTS Descriptive statistics (mean ± standard deviation). RESULTS Biopsy was performed a mean of 9 days (range 5-19 days) after HP [1-13C]pyruvate MRI. Three biopsies were normal, one showed low-grade fibrosis and one showed moderate microvascular inflammation. All had stable functioning allografts with eGFR > 60 mL/min/1.73 m2 and normal uPCR. One participant who did not undergo biopsy had reduced eGFR of 49 mL/min/1.73 m2 and elevated uPCR. The mean lactate-to-pyruvate ratio was 0.373 in participants with normal findings (n = 3) and 0.552 in participants with abnormal findings (n = 2). The lactate-to-pyruvate ratio was highest (0.847) in the participant with reduced eGFR and elevated uPRC. Native non-tumor bearing kidneys had a mean lactate-to-pyruvate ratio of 0.309. DATA CONCLUSION Stable allografts with normal findings at biopsy showed lactate-to-pyruvate ratios similar to native non-tumor bearing kidneys, whereas allografts with abnormal findings showed higher lactate-to-pyruvate ratios.
Collapse
Affiliation(s)
- Xiaoxi Liu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Ying-Chieh Lai
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan
| | - Di Cui
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Shiang-Cheng Kung
- Department of Medicine, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Meyeon Park
- Department of Medicine, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Laszik Zoltan
- Department of Pathology, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Peder E.Z. Larson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Zhen J. Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
2
|
Liu X, Lai YC, Cui D, Kung SC, Park M, Laszik Z, Larson PEZ, Wang ZJ. Initial Experience of Metabolic Imaging With Hyperpolarized [1- 13C]pyruvate MRI in Kidney Transplant Patients. J Magn Reson Imaging 2024. [PMID: 39239784 DOI: 10.1002/jmri.29580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Kidney transplant is the treatment of choice for patients with end-stage renal disease. Early detection of allograft injury is important to delay or prevent irreversible damage. PURPOSE To investigate the feasibility of hyperpolarized (HP) [1-13C]pyruvate MRI for assessing kidney allograft metabolism. STUDY TYPE Prospective. SUBJECTS Six participants (mean age, 45.2 ± 12.4 years, two females) scheduled for kidney allograft biopsy and five patients (mean age, 59.6 ± 10.4 years, two females) with renal cell carcinoma (RCC). FIELD STRENGTH/SEQUENCE Three Tesla, T2-weighted fast spin echo, multi-echo gradient echo, single shot diffusion-weighted echo-planar imaging, and time-resolved HP 13C metabolite-selective imaging. ASSESSMENT Five of the six kidney allograft participants underwent biopsy after MRI. Estimated glomerular filtration rate (eGFR) and urine protein-to-creatine ratio (uPCR) were collected within 4 weeks of MRI. Kidney metabolism was quantified from HP [1-13C]pyruvate MRI using the lactate-to-pyruvate ratio in allograft kidneys and non-tumor bearing kidneys from RCC patients. STATISTICAL TESTS Descriptive statistics (mean ± SD). RESULTS Biopsy was performed a mean of 9 days (range 5-19 days) after HP [1-13C]pyruvate MRI. Three biopsies were normal, one showed low-grade fibrosis and one showed moderate microvascular inflammation. All had stable functioning allografts with eGFR >60 mL/min/1.73 m2 and normal uPCR. One participant who did not undergo biopsy had reduced eGFR of 49 mL/min/1.73 m2 and elevated uPCR. The mean lactate-to-pyruvate ratio was 0.373 in participants with normal findings (N = 3) and 0.552 in participants with abnormal findings (N = 2). The lactate-to-pyruvate ratio was highest (0.847) in the participant with reduced eGFR and elevated uPRC. Native non-tumor bearing kidneys had a mean lactate-to-pyruvate ratio of 0.309. DATA CONCLUSION Stable allografts with normal findings at biopsy showed lactate-to-pyruvate ratios similar to native non-tumor bearing kidneys, whereas allografts with abnormal findings showed higher lactate-to-pyruvate ratios. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Xiaoxi Liu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Ying-Chieh Lai
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Di Cui
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Shiang-Cheng Kung
- Department of Medicine, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Meyeon Park
- Department of Medicine, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Zoltan Laszik
- Department of Pathology, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
3
|
Carswell G, Chamberlin J, Bennett BD, Bushel PR, Chorley BN. Persistent gene expression and DNA methylation alterations linked to carcinogenic effects of dichloroacetic acid. Front Oncol 2024; 14:1389634. [PMID: 38764585 PMCID: PMC11099211 DOI: 10.3389/fonc.2024.1389634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 05/21/2024] Open
Abstract
Background Mechanistic understanding of transient exposures that lead to adverse health outcomes will enhance our ability to recognize biological signatures of disease. Here, we measured the transcriptomic and epigenomic alterations due to exposure to the metabolic reprogramming agent, dichloroacetic acid (DCA). Previously, we showed that exposure to DCA increased liver tumor incidence in B6C3F1 mice after continuous or early life exposures significantly over background level. Methods Using archived formalin-fixed liver samples, we utilized modern methodologies to measure gene expression and DNA methylation levels to link to previously generated phenotypic measures. Gene expression was measured by targeted RNA sequencing (TempO-seq 1500+ toxicity panel: 2754 total genes) in liver samples collected from 10-, 32-, 57-, and 78-week old mice exposed to deionized water (controls), 3.5 g/L DCA continuously in drinking water ("Direct" group), or DCA for 10-, 32-, or 57-weeks followed by deionized water until sample collection ("Stop" groups). Genome-scaled alterations in DNA methylation were measured by Reduced Representation Bisulfite Sequencing (RRBS) in 78-week liver samples for control, Direct, 10-week Stop DCA exposed mice. Results Transcriptomic changes were most robust with concurrent or adjacent timepoints after exposure was withdrawn. We observed a similar pattern with DNA methylation alterations where we noted attenuated differentially methylated regions (DMRs) in the 10-week Stop DCA exposure groups compared to the Direct group at 78-weeks. Gene pathway analysis indicated cellular effects linked to increased oxidative metabolism, a primary mechanism of action for DCA, closer to exposure windows especially early in life. Conversely, many gene signatures and pathways reversed patterns later in life and reflected more pro-tumorigenic patterns for both current and prior DCA exposures. DNA methylation patterns correlated to early gene pathway perturbations, such as cellular signaling, regulation and metabolism, suggesting persistence in the epigenome and possible regulatory effects. Conclusion Liver metabolic reprogramming effects of DCA interacted with normal age mechanisms, increasing tumor burden with both continuous and prior DCA exposure in the male B6C3F1 rodent model.
Collapse
Affiliation(s)
- Gleta Carswell
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - John Chamberlin
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Brian D. Bennett
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, United States
| | - Pierre R. Bushel
- Massive Genome Informatics Group, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, United States
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, United States
| | - Brian N. Chorley
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| |
Collapse
|
4
|
Liu X, Li J, Huang Q, Jin M, Huang G. Ginsenoside Rh2 shifts tumor metabolism from aerobic glycolysis to oxidative phosphorylation through regulating the HIF1-α/PDK4 axis in non-small cell lung cancer. Mol Med 2024; 30:56. [PMID: 38671369 PMCID: PMC11055298 DOI: 10.1186/s10020-024-00813-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Ginsenoside Rh2 (G-Rh2), a steroidal compound extracted from roots of ginseng, has been extensively studied in tumor therapy. However, its specific regulatory mechanism in non-small cell lung cancer (NSCLC) is not well understood. Pyruvate dehydrogenase kinase 4 (PDK4), a central regulator of cellular energy metabolism, is highly expressed in various malignant tumors. We investigated the impact of G-Rh2 on the malignant progression of NSCLC and how it regulated PDK4 to influence tumor aerobic glycolysis and mitochondrial function. METHOD We examined the inhibitory effect of G-Rh2 on NSCLC through I proliferation assay, migration assay and flow cytometry in vitro. Subsequently, we verified the ability of G-Rh2 to inhibit tumor growth and metastasis by constructing subcutaneous tumor and metastasis models in nude mice. Proteomics analysis was conducted to analyze the action pathways of G-Rh2. Additionally, we assessed glycolysis and mitochondrial function using seahorse, PET-CT, Western blot, and RT-qPCR. RESULT Treatment with G-Rh2 significantly inhibited tumor proliferation and migration ability both in vitro and in vivo. Furthermore, G-Rh2 inhibited the tumor's aerobic glycolytic capacity, including glucose uptake and lactate production, through the HIF1-α/PDK4 pathway. Overexpression of PDK4 demonstrated that G-Rh2 targeted the inhibition of PDK4 expression, thereby restoring mitochondrial function, promoting reactive oxygen species (ROS) accumulation, and inducing apoptosis. When combined with sodium dichloroacetate, a PDK inhibitor, it complemented the inhibitory capacity of PDKs, acting synergistically as a detoxifier. CONCLUSION G-Rh2 could target and down-regulate the expression of HIF-1α, resulting in decreased expression of glycolytic enzymes and inhibition of aerobic glycolysis in tumors. Additionally, by directly targeting mitochondrial PDK, it elevated mitochondrial oxidative phosphorylation and enhanced ROS accumulation, thereby promoting tumor cells to undergo normal apoptotic processes.
Collapse
Affiliation(s)
- Xiyu Liu
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China
| | - Jingjing Li
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China.
| | - Gang Huang
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, P.R. China.
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Pudong New Area, 201318, Shanghai, China.
| |
Collapse
|
5
|
Li Y, Xie Z, Lei X, Yang X, Huang S, Yuan W, Deng X, Wang Z, Tang G. Recent advances in pyruvate dehydrogenase kinase inhibitors: Structures, inhibitory mechanisms and biological activities. Bioorg Chem 2024; 144:107160. [PMID: 38301426 DOI: 10.1016/j.bioorg.2024.107160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/03/2024]
Abstract
Metabolism is reprogrammed in a variety of cancer cells to ensure their rapid proliferation. Cancer cells prefer to utilize glycolysis to produce energy as well as to provide large amounts of precursors for their division. In this process, cancer cells inhibit the activity of pyruvate dehydrogenase complex (PDC) by upregulating the expression of pyruvate dehydrogenase kinases (PDKs). Inhibiting the activity of PDKs in cancer cells can effectively block this metabolic transition in cancer cells, while also activating mitochondrial oxidative metabolism and promoting apoptosis of cancer cells. To this day, the study of PDKs inhibitors has become one of the research hotspots in the field of medicinal chemistry. Novel structures targeting PDKs are constantly being discovered, and some inhibitors have entered the clinical research stage. Here, we reviewed the research progress of PDKs inhibitors in recent years and classified them according to the PDKs binding sites they acted on, aiming to summarize the structural characteristics of inhibitors acting on different binding sites and explore their clinical application value. Finally, the shortcomings of some PDKs inhibitors and the further development direction of PDKs inhibitors are discussed.
Collapse
Affiliation(s)
- Yiyang Li
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyan Yang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Sheng Huang
- Jiuzhitang Co., Ltd, Changsha, Hunan 410007, China
| | - Weixi Yuan
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
6
|
Jiang D, Guo J, Liu Y, Li W, Lu D. Glycolysis: an emerging regulator of osteoarthritis. Front Immunol 2024; 14:1327852. [PMID: 38264652 PMCID: PMC10803532 DOI: 10.3389/fimmu.2023.1327852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Osteoarthritis (OA) has been a leading cause of disability in the elderly and there remains a lack of effective therapeutic approaches as the mechanisms of pathogenesis and progression have yet to be elucidated. As OA progresses, cellular metabolic profiles and energy production are altered, and emerging metabolic reprogramming highlights the importance of specific metabolic pathways in disease progression. As a crucial part of glucose metabolism, glycolysis bridges metabolic and inflammatory dysfunctions. Moreover, the glycolytic pathway is involved in different areas of metabolism and inflammation, and is associated with a variety of transcription factors. To date, it has not been fully elucidated whether the changes in the glycolytic pathway and its associated key enzymes are associated with the onset or progression of OA. This review summarizes the important role of glycolysis in mediating cellular metabolic reprogramming in OA and its role in inducing tissue inflammation and injury, with the aim of providing further insights into its pathological functions and proposing new targets for the treatment of OA.
Collapse
Affiliation(s)
- Dingming Jiang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingquan Liu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenxin Li
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Hangzhou Linping District Nanyuan Street Community Health Center, Hangzhou, China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Pearl PL, Tokatly Latzer I, Lee HHC, Rotenberg A. New Therapeutic Approaches to Inherited Metabolic Pediatric Epilepsies. Neurology 2023; 101:124-133. [PMID: 36878704 PMCID: PMC10382274 DOI: 10.1212/wnl.0000000000207133] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/17/2023] [Indexed: 03/08/2023] Open
Abstract
Treatment options for inherited metabolic epilepsies are rapidly expanding with advances in molecular biology and the genomic revolution. Traditional dietary and nutrient modification and inhibitors or enhancers of protein and enzyme function, the mainstays of therapy, are undergoing continuous revisions to increase biological activity and reduce toxicity. Enzyme replacement and gene replacement and editing hold promise for genetically targeted treatment and cures. Molecular, imaging, and neurophysiologic biomarkers are emerging as key indicators of disease pathophysiology, severity, and response to therapy.
Collapse
Affiliation(s)
- Phillip L Pearl
- From the Department of Neurology (P.L.P., I.T.L., H.H.C.L., A.R.), Boston Children's Hospital, Harvard Medical School, Boston, MA.
| | - Itay Tokatly Latzer
- From the Department of Neurology (P.L.P., I.T.L., H.H.C.L., A.R.), Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Henry H C Lee
- From the Department of Neurology (P.L.P., I.T.L., H.H.C.L., A.R.), Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Alexander Rotenberg
- From the Department of Neurology (P.L.P., I.T.L., H.H.C.L., A.R.), Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
8
|
Wang J, Zhang Y, Luo Y, Liu ML, Niu W, Li ZC, Zhang B. PDK1 upregulates PINK1-mediated pulmonary endothelial cell mitophagy during hypoxia-induced pulmonary vascular remodeling. Mol Biol Rep 2023; 50:5585-5596. [PMID: 37162681 DOI: 10.1007/s11033-023-08428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/04/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Hypoxic pulmonary hypertension (HPH) is a complication of lung diseases with pulmonary vascular remodeling, although the underlying molecular mechanisms have not been fully elucidated. This study investigated the underlying molecular events by using a rat HPH model and primary pulmonary microvascular endothelial cells (PMVECs). METHODS AND RESULTS This study first established a rat HPH model and cultured PMVECs for transmission electron microscopic analysis and manipulation of 3-phosphoinositide-dependent protein kinase 1 (PDK1) or phosphatase and tensin homolog-induced kinase 1 (PINK1) expression in vitro. After that, the cell viability was assessed and the expression of different proteins was assayed using cell viability and western blot assays, respectively. Reactive oxygen species production, apoptosis, NLR family pyrin domain containing 3 (NLRP3) expression, and the levels of interleukin (IL)-1β, IL-6, and IL-8 were also assessed, while the interaction of PDK1 and PINK1 was determined using co-immunoprecipitation/western blot assays. Hypoxia induced mitophagy in the PMVECs and upregulated PINK1/Parkin expression, whereas knockdown of PINK1 expression under hypoxic conditions inhibited cell proliferation but induced endothelial cell apoptosis in vitro, decreased reactive oxygen species production and NLRP3 expression, and reduced the levels of inflammatory factors in PMVECs. However, hypoxia induced PDK1 expression, whereas knockdown of PDK1 downregulated PINK1 expression. Furthermore, treatment of the model rats with the PDK1 inhibitor dichloroacetate (DCA) was able to decrease PINK1 expression. In addition, the PDK1 and PINK1 proteins could interact with each other in the mitochondria of PMVECs to regulate the cell viability. CONCLUSIONS This study revealed that PDK1 induced PMVEC proliferation but inhibited their apoptosis to participate in pulmonary vascular remodeling, ultimately leading to HPH through regulation of PINK1-mediated mitophagy signaling. Therefore, PINK1 is a novel therapeutic target for the control of HPH.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Yue Zhang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Ying Luo
- Department of Physiology and Pathophysiology, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Man Ling Liu
- Department of Physiology and Pathophysiology, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Wen Niu
- Department of Physiology and Pathophysiology, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Zhi Chao Li
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China.
- Department of Physiology and Pathophysiology, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, Shaanxi, China.
| | - Bo Zhang
- Department of Physiology and Pathophysiology, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
9
|
Tokatly Latzer I, Pearl PL. Treatment of neurometabolic epilepsies: Overview and recent advances. Epilepsy Behav 2023; 142:109181. [PMID: 37001467 DOI: 10.1016/j.yebeh.2023.109181] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/11/2023] [Accepted: 03/12/2023] [Indexed: 05/08/2023]
Abstract
The rarity and heterogeneity of neurometabolic diseases make it challenging to reach evidence-based principles for their specific treatments. Indeed, current treatments for many of these diseases remain symptomatic and supportive. However, an ongoing scientific and medical revolution has led to dramatic breakthroughs in molecular sciences and genetics, revealing precise pathophysiologic mechanisms. Accordingly, this has led to significant progress in the development of novel therapeutic approaches aimed at treating epilepsy resulting from these conditions, as well as their other manifestations. We overview recent notable treatment advancements, from vitamins, trace minerals, and diets to unique medications targeting the elemental pathophysiology at a molecular or cellular level, including enzyme replacement therapy, enzyme enhancing therapy, antisense oligonucleotide therapy, stem cell transplantation, and gene therapy.
Collapse
Affiliation(s)
- Itay Tokatly Latzer
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Pecoraro C, De Franco M, Carbone D, Bassani D, Pavan M, Cascioferro S, Parrino B, Cirrincione G, Dall'Acqua S, Moro S, Gandin V, Diana P. 1,2,4-Amino-triazine derivatives as pyruvate dehydrogenase kinase inhibitors: Synthesis and pharmacological evaluation. Eur J Med Chem 2023; 249:115134. [PMID: 36709650 DOI: 10.1016/j.ejmech.2023.115134] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/08/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
Among the different hallmarks of cancer, deregulation of cellular metabolism turned out to be an essential mechanism in promoting cancer resistance and progression. The pyruvate dehydrogenase kinases (PDKs) are well known as key regulators in cells metabolic process and their activity was found to be overexpressed in different metabolic alerted types of cancer, including the high aggressive pancreatic ductal adenocarcinoma (PDAC). To date few PDK inhibitors have been reported, and the different molecules developed are characterized by structural chemical diversity. In an attempt to find novel classes of potential PDK inhibitors, the molecular hybridization approach, which combine two or more active scaffolds in a single structure, was employed. Herein we report the synthesis and the pharmacological evaluation of the novel hybrid molecules, characterized by the fusion of three different pharmacophoric sub-units such as 1,2,4-amino triazines, 7-azaindoles and indoles, in a single structure. The synthesized derivatives demonstrated a promising ability in hampering the enzymatic activity of PDK1 and 4, further confirmed by docking studies. Interestingly, these derivatives retained a strong antiproliferative activity against pancreatic cancer cells either in 2D and 3D models. Mechanistic studies in highly aggressive PDAC cells confirmed their ability to hamper PDK axis and to induce cancer cell death by apoptosis. Moreover, in vivo translational studies in a murine syngeneic solid tumor model confirmed the ability of the most representative compounds to target the PDK system and highlight the ability to reduce the tumor growth without inducing substantial body weight changes in the treated mice.
Collapse
Affiliation(s)
- Camilla Pecoraro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123, Palermo, Italy
| | - Michele De Franco
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131, Padova, Italy
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123, Palermo, Italy
| | - Davide Bassani
- Department of Pharmaceutical and Pharmacological Sciences, Molecular Modeling Section (MMS), University of Padova, via F. Marzolo 5, 35131, Padova, Italy
| | - Matteo Pavan
- Department of Pharmaceutical and Pharmacological Sciences, Molecular Modeling Section (MMS), University of Padova, via F. Marzolo 5, 35131, Padova, Italy
| | - Stella Cascioferro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123, Palermo, Italy
| | - Barbara Parrino
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123, Palermo, Italy
| | - Girolamo Cirrincione
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123, Palermo, Italy
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131, Padova, Italy
| | - Stefano Moro
- Department of Pharmaceutical and Pharmacological Sciences, Molecular Modeling Section (MMS), University of Padova, via F. Marzolo 5, 35131, Padova, Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, 35131, Padova, Italy.
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123, Palermo, Italy.
| |
Collapse
|
11
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
12
|
Zare S, Ramezani Z, Ghadiri AA, Fereidoonnezhad M. Synthesis of N‐(2‐(tert‐Butylamino)‐2‐oxoethyl)‐2,2‐dichloro‐N‐aryl(alkyl)acetamides as Anticancer Agents: Molecular Modeling and Biological Evaluations. ChemistrySelect 2023. [DOI: 10.1002/slct.202203931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Somayeh Zare
- Cancer Research Center Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Medicinal Chemistry School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Zahra Ramezani
- Cancer Research Center Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Medicinal Chemistry Faculty of Pharmacy Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Ata A. Ghadiri
- Department of Immunology School of Medicine Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Masood Fereidoonnezhad
- Cancer Research Center Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
- Department of Medicinal Chemistry Faculty of Pharmacy Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| |
Collapse
|
13
|
Lee SH, Choi BY, Kho AR, Hong DK, Kang BS, Park MK, Lee SH, Choi HC, Song HK, Suh SW. Combined Treatment of Dichloroacetic Acid and Pyruvate Increased Neuronal Survival after Seizure. Nutrients 2022; 14:4804. [PMID: 36432491 PMCID: PMC9698956 DOI: 10.3390/nu14224804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
During seizure activity, glucose and Adenosine triphosphate (ATP) levels are significantly decreased in the brain, which is a contributing factor to seizure-induced neuronal death. Dichloroacetic acid (DCA) has been shown to prevent cell death. DCA is also known to be involved in adenosine triphosphate (ATP) production by activating pyruvate dehydrogenase (PDH), a gatekeeper of glucose oxidation, as a pyruvate dehydrogenase kinase (PDK) inhibitor. To confirm these findings, in this study, rats were given a per oral (P.O.) injection of DCA (100 mg/kg) with pyruvate (50 mg/kg) once per day for 1 week starting 2 h after the onset of seizures induced by pilocarpine administration. Neuronal death and oxidative stress were assessed 1 week after seizure to determine if the combined treatment of pyruvate and DCA increased neuronal survival and reduced oxidative damage in the hippocampus. We found that the combined treatment of pyruvate and DCA showed protective effects against seizure-associated hippocampal neuronal cell death compared to the vehicle-treated group. Treatment with combined pyruvate and DCA after seizure may have a therapeutic effect by increasing the proportion of pyruvate converted to ATP. Thus, the current research demonstrates that the combined treatment of pyruvate and DCA may have therapeutic potential in seizure-induced neuronal death.
Collapse
Affiliation(s)
- Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Bo Young Choi
- Department of Physical Education, Hallym University, Chuncheon 24252, Korea
- Institute of Sports Science, Hallym University, Chuncheon 24252, Korea
| | - A Ra Kho
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Si Hyun Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Hui Chul Choi
- College of Medicine, Neurology, Hallym University, Chuncheon 24252, Korea
- Hallym Institute of Epilepsy Research, Hallym University, Chuncheon 24252, Korea
| | - Hong Ki Song
- College of Medicine, Neurology, Hallym University, Chuncheon 24252, Korea
- Hallym Institute of Epilepsy Research, Hallym University, Chuncheon 24252, Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Hallym Institute of Epilepsy Research, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
14
|
Chen G, She W, Yu C, Rouzi T, Li X, Ma L, Zhang N, Jiang H, Liu X, Wu J, Wang Q, Shen H, Zhou F. A novel organic arsenic derivative MZ2 remodels metabolism and triggers mtROS-mediated apoptosis in acute myeloid leukemia. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04333-2. [PMID: 36056952 DOI: 10.1007/s00432-022-04333-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Acute myeloid leukemia (AML) is one of the most common neoplasms in adults, and it is difficult to achieve satisfactory results with conventional drugs. Here, we synthesized a novel organic arsenic derivative MZ2 and evaluated its ability to remodel energy metabolism to achieve anti-leukemia. METHODS MZ2 was characterized by the average 1-min full mass spectra analysis. Biological methods such as Western blot, qPCR, flow cytometry and confocal microscopy were used to assess the mode and mechanism of MZ2-induced death. The in vivo efficacy of MZ2 was assessed by constructing a patient-derived xenograft (PDX) AML model. RESULTS Unlike the precursor organic arsenical Z2, MZ2 can effectively reduce the level of aerobic glycolysis. Our in-depth found that MZ2 inhibited the expression of PDK2 in a dose-dependent manner and did not affect the expression of LDHA, another key enzyme of the glycolytic pathway. MZ2 reconstituted energy metabolism to induce the generation of mitochondrial ROS (mtROS) and then triggerd intrinsic apoptosis pathway. We also assessed whether MZ2 generates autophagy and results showed that MZ2 can induce autophagy of AML cells, which may be associated with the precursor organic arsenic drug. In vivo, MZ2 effectively attenuated leukemia progression in mice, and immunohistochemical results suggested its PDK2 inhibitory effect. CONCLUSION In summary, the novel organic arsine derivative MZ2 exhibited excellent anti-tumor effects in acute myeloid leukemia, which may provide a potential strategy for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Guopeng Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Wenyan She
- College of Chemistry and Molecular Science, Wuhan University, Wuhan, 430072, Hubei, China
| | - Chaochao Yu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Tuerxunayi Rouzi
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinqi Li
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Linlu Ma
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Nan Zhang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Hongqiang Jiang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiaoyan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Jinxian Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Qian Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Hui Shen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
15
|
Contribution of Mendelian Disorders in a Population-Based Pediatric Neurodegeneration Cohort. J Pediatr 2022; 248:89-93. [PMID: 35577121 DOI: 10.1016/j.jpeds.2022.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To evaluate Mendelian causes of neurodegenerative disorders in a cohort of pediatric patients. STUDY DESIGN Patients enrolled in the Center for Applied Genomics Biobank at the Children's Hospital of Philadelphia with neurodegenerative symptoms were identified using an algorithm that consisted of including and excluding selected International Classification of Diseases, 9th and 10th edition codes. A manual chart review was then performed to abstract detailed clinical information. RESULTS Of approximately 100 000 patients enrolled in the Center for Applied Genomics Biobank, 76 had a neurodegenerative phenotype. After chart review, 7 patients were excluded. Of the remaining 69 patients, 42 had a genetic diagnosis (60.9%) and 27 were undiagnosed (39.1%). There were 32 unique disorders. Common diagnoses included Rett syndrome, mitochondrial disorders, and neuronal ceroid lipofuscinoses. CONCLUSIONS The disorders encountered in our cohort demonstrate the diverse diseases and pathophysiology that contribute to pediatric neurodegeneration. Establishing a diagnosis often informed clinical management, although curative treatment options are lacking. Many patients who underwent genetic evaluation remained undiagnosed, highlighting the importance of continued research efforts in this field.
Collapse
|
16
|
Lavorato M, Nakamaru-Ogiso E, Mathew ND, Herman E, Shah NK, Haroon S, Xiao R, Seiler C, Falk MJ. Dichloroacetate improves mitochondrial function, physiology, and morphology in FBXL4 disease models. JCI Insight 2022; 7:156346. [PMID: 35881484 PMCID: PMC9462489 DOI: 10.1172/jci.insight.156346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Pathogenic variants in the human F-box and leucine-rich repeat protein 4 (FBXL4) gene result in an autosomal recessive, multisystemic, mitochondrial disorder involving variable mitochondrial depletion and respiratory chain complex deficiencies with lactic acidemia. As no FDA-approved effective therapies for this disease exist, we sought to characterize translational C. elegans and zebrafish animal models, as well as human fibroblasts, to study FBXL4–/– disease mechanisms and identify preclinical therapeutic leads. Developmental delay, impaired fecundity and neurologic and/or muscular activity, mitochondrial dysfunction, and altered lactate metabolism were identified in fbxl-1(ok3741) C. elegans. Detailed studies of a PDHc activator, dichloroacetate (DCA), in fbxl-1(ok3741)C. elegans demonstrated its beneficial effects on fecundity, neuromotor activity, and mitochondrial function. Validation studies were performed in fbxl4sa12470 zebrafish larvae and in FBXL4–/– human fibroblasts; they showed DCA efficacy in preventing brain death, impairment of neurologic and/or muscular function, mitochondrial biochemical dysfunction, and stress-induced morphologic and ultrastructural mitochondrial defects. These data demonstrate that fbxl-1(ok3741) C. elegans and fbxl4sa12470 zebrafish provide robust translational models to study mechanisms and identify preclinical therapeutic candidates for FBXL4–/– disease. Furthermore, DCA is a lead therapeutic candidate with therapeutic benefit on diverse aspects of survival, neurologic and/or muscular function, and mitochondrial physiology that warrants rigorous clinical trial study in humans with FBXL4–/– disease.
Collapse
Affiliation(s)
- Manuela Lavorato
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Eiko Nakamaru-Ogiso
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Neal D Mathew
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Elizabeth Herman
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Nina K Shah
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Suraiya Haroon
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Rui Xiao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, United States of America
| | - Christoph Seiler
- Aquatics Core Facility, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| |
Collapse
|
17
|
Karissa P, Simpson T, Dawson SP, Low TY, Tay SH, Nordin FDA, Zain SM, Lee PY, Pung YF. Comparison Between Dichloroacetate and Phenylbutyrate Treatment for Pyruvate Dehydrogenase Deficiency. Br J Biomed Sci 2022; 79:10382. [PMID: 35996497 PMCID: PMC9302545 DOI: 10.3389/bjbs.2022.10382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022]
Abstract
Pyruvate dehydrogenase (PDH) deficiency is caused by a number of pathogenic variants and the most common are found in the PDHA1 gene. The PDHA1 gene encodes one of the subunits of the PDH enzyme found in a carbohydrate metabolism pathway involved in energy production. Pathogenic variants of PDHA1 gene usually impact the α-subunit of PDH causing energy reduction. It potentially leads to increased mortality in sufferers. Potential treatments for this disease include dichloroacetate and phenylbutyrate, previously used for other diseases such as cancer and maple syrup urine disease. However, not much is known about their efficacy in treating PDH deficiency. Effective treatment for PDH deficiency is crucial as carbohydrate is needed in a healthy diet and rice is the staple food for a large portion of the Asian population. This review analysed the efficacy of dichloroacetate and phenylbutyrate as potential treatments for PDH deficiency caused by PDHA1 pathogenic variants. Based on the findings of this review, dichloroacetate will have an effect on most PDHA1 pathogenic variant and can act as a temporary treatment to reduce the lactic acidosis, a common symptom of PDH deficiency. Phenylbutyrate can only be used on patients with certain pathogenic variants (p.P221L, p.R234G, p.G249R, p.R349C, p.R349H) on the PDH protein. It is hoped that the review would provide an insight into these treatments and improve the quality of lives for patients with PDH deficiency.
Collapse
Affiliation(s)
- Patricia Karissa
- Division of Biomedical Science, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Timothy Simpson
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Simon P Dawson
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sook Hui Tay
- Division of Biomedical Science, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | | | - Shamsul Mohd Zain
- Department of Pharmacology, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yuh-Fen Pung
- Division of Biomedical Science, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| |
Collapse
|
18
|
Phase II study of dichloroacetate, an inhibitor of pyruvate dehydrogenase, in combination with chemoradiotherapy for unresected, locally advanced head and neck squamous cell carcinoma. Invest New Drugs 2022; 40:622-633. [DOI: 10.1007/s10637-022-01235-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/11/2022] [Indexed: 12/14/2022]
|
19
|
Simonsen D, Cady N, Zhang C, Shrode RL, McCormick ML, Spitz DR, Chimenti MS, Wang K, Mangalam A, Lehmler HJ. The Effects of Benoxacor on the Liver and Gut Microbiome of C57BL/6 Mice. Toxicol Sci 2022; 186:102-117. [PMID: 34850242 PMCID: PMC9019840 DOI: 10.1093/toxsci/kfab142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The toxicity of many "inert" ingredients of pesticide formulations, such as safeners, is poorly characterized, despite evidence that humans may be exposed to these chemicals. Analysis of ToxCast data for dichloroacetamide safeners with the ToxPi tool identified benoxacor as the safener with the highest potential for toxicity, especially liver toxicity. Benoxacor was subsequently administered to mice via oral gavage for 3 days at concentrations of 0, 0.5, 5, and 50 mg/kg bodyweight (b.w.). Bodyweight-adjusted liver and testes weights were significantly increased in the 50 mg/kg b.w. group. There were no overt pathologies in either the liver or the intestine. 16S rRNA analysis of the cecal microbiome revealed no effects of benoxacor on α- or β-diversity; however, changes were observed in the abundance of certain bacteria. RNAseq analysis identified 163 hepatic genes affected by benoxacor exposure. Benoxacor exposure expressed a gene regulation profile similar to dichloroacetic acid and the fungicide sedaxane. Metabolomic analysis identified 9 serum and 15 liver metabolites that were affected by benoxacor exposure, changes that were not significant after correcting for multiple comparisons. The activity of antioxidant enzymes was not altered by benoxacor exposure. In vitro metabolism studies with liver microsomes and cytosol from male mice demonstrated that benoxacor is enantioselectively metabolized by cytochrome P450 enzymes, carboxylesterases, and glutathione S-transferases. These findings suggest that the minor toxic effects of benoxacor may be due to its rapid metabolism to toxic metabolites, such as dichloroacetic acid. This result challenges the assumption that inert ingredients of pesticide formulations are safe.
Collapse
Affiliation(s)
- Derek Simonsen
- Department of Occupational and Environmental Health, The University of Iowa, Iowa City, Iowa 52242, USA
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa 52242, USA
- IIHR Hydroscience and Engineering, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Nicole Cady
- Department of Pathology, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Chunyun Zhang
- Department of Occupational and Environmental Health, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Rachel L Shrode
- Department of Informatics, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Kai Wang
- Department of Biostatistics, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Ashutosh Mangalam
- Department of Pathology, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, The University of Iowa, Iowa City, Iowa 52242, USA
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa 52242, USA
- IIHR Hydroscience and Engineering, The University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
20
|
The metabolism of cells regulates their sensitivity to NK cells depending on p53 status. Sci Rep 2022; 12:3234. [PMID: 35217717 PMCID: PMC8881467 DOI: 10.1038/s41598-022-07281-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Leukemic cells proliferate faster than non-transformed counterparts. This requires them to change their metabolism to adapt to their high growth. This change can stress cells and facilitate recognition by immune cells such as cytotoxic lymphocytes, which express the activating receptor Natural Killer G2-D (NKG2D). The tumor suppressor gene p53 regulates cell metabolism, but its role in the expression of metabolism-induced ligands, and subsequent recognition by cytotoxic lymphocytes, is unknown. We show here that dichloroacetate (DCA), which induces oxidative phosphorylation (OXPHOS) in tumor cells, induces the expression of such ligands, e.g. MICA/B, ULBP1 and ICAM-I, by a wtp53-dependent mechanism. Mutant or null p53 have the opposite effect. Conversely, DCA sensitizes only wtp53-expressing cells to cytotoxic lymphocytes, i.e. cytotoxic T lymphocytes and NK cells. In xenograft in vivo models, DCA slows down the growth of tumors with low proliferation. Treatment with DCA, monoclonal antibodies and NK cells also decreased tumors with high proliferation. Treatment of patients with DCA, or a biosimilar drug, could be a clinical option to increase the effectiveness of CAR T cell or allogeneic NK cell therapies.
Collapse
|
21
|
Parkin ET, Hammond JE, Owens L, Hodges MD. The orphan drug dichloroacetate reduces amyloid beta-peptide production whilst promoting non-amyloidogenic proteolysis of the amyloid precursor protein. PLoS One 2022; 17:e0255715. [PMID: 35025874 PMCID: PMC8757967 DOI: 10.1371/journal.pone.0255715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/20/2021] [Indexed: 11/18/2022] Open
Abstract
The amyloid cascade hypothesis proposes that excessive accumulation of amyloid beta-peptides is the initiating event in Alzheimer’s disease. These neurotoxic peptides are generated from the amyloid precursor protein via sequential cleavage by β- and γ-secretases in the ’amyloidogenic’ proteolytic pathway. Alternatively, the amyloid precursor protein can be processed via the ’non-amyloidogenic’ pathway which, through the action of the α-secretase a disintegrin and metalloproteinase (ADAM) 10, both precludes amyloid beta-peptide formation and has the additional benefit of generating a neuroprotective soluble amyloid precursor protein fragment, sAPPα. In the current study, we investigated whether the orphan drug, dichloroacetate, could alter amyloid precursor protein proteolysis. In SH-SY5Y neuroblastoma cells, dichloroacetate enhanced sAPPα generation whilst inhibiting β–secretase processing of endogenous amyloid precursor protein and the subsequent generation of amyloid beta-peptides. Over-expression of the amyloid precursor protein partly ablated the effect of dichloroacetate on amyloidogenic and non-amyloidogenic processing whilst over-expression of the β-secretase only ablated the effect on amyloidogenic processing. Similar enhancement of ADAM-mediated amyloid precursor protein processing by dichloroacetate was observed in unrelated cell lines and the effect was not exclusive to the amyloid precursor protein as an ADAM substrate, as indicated by dichloroacetate-enhanced proteolysis of the Notch ligand, Jagged1. Despite altering proteolysis of the amyloid precursor protein, dichloroacetate did not significantly affect the expression/activity of α-, β- or γ-secretases. In conclusion, dichloroacetate can inhibit amyloidogenic and promote non-amyloidogenic proteolysis of the amyloid precursor protein. Given the small size and blood-brain-barrier permeability of the drug, further research into its mechanism of action with respect to APP proteolysis may lead to the development of therapies for slowing the progression of Alzheimer’s disease.
Collapse
Affiliation(s)
- Edward T. Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
- * E-mail:
| | - Jessica E. Hammond
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Lauren Owens
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Matthew D. Hodges
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| |
Collapse
|
22
|
Zhao X, Li S, Mo Y, Li R, Huang S, Zhang A, Ni X, Dai Q, Wang J. DCA Protects against Oxidation Injury Attributed to Cerebral Ischemia-Reperfusion by Regulating Glycolysis through PDK2-PDH-Nrf2 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5173035. [PMID: 34712383 PMCID: PMC8548159 DOI: 10.1155/2021/5173035] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/06/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022]
Abstract
Cerebral ischemic stroke (IS) is still a difficult problem to be solved; energy metabolism failure is one of the main factors causing mitochondrion dysfunction and oxidation stress damage within the pathogenesis of cerebral ischemia, which produces considerable reactive oxygen species (ROS) and opens the blood-brain barrier. Dichloroacetic acid (DCA) can inhibit pyruvate dehydrogenase kinase (PDK). Moreover, DCA has been indicated with the capability of increasing mitochondrial pyruvate uptake and promoting oxidation of glucose in the course of glycolysis, thereby improving the activity of pyruvate dehydrogenase (PDH). As a result, pyruvate flow is promoted into the tricarboxylic acid cycle to expedite ATP production. DCA has a protective effect on IS and brain ischemia/reperfusion (I/R) injury, but the specific mechanism remains unclear. This study adopted a transient middle cerebral artery occlusion (MCAO) mouse model for simulating IS and I/R injury in mice. We investigated the mechanism by which DCA regulates glycolysis and protects the oxidative damage induced by I/R injury through the PDK2-PDH-Nrf2 axis. As indicated from the results of this study, DCA may improve glycolysis, reduce oxidative stress and neuronal death, damage the blood-brain barrier, and promote the recovery of oxidative metabolism through inhibiting PDK2 and activating PDH. Additionally, DCA noticeably elevated the neurological score and reduced the infarct volume, brain water content, and necrotic neurons. Moreover, as suggested from the results, DCA elevated the content of Nrf2 as well as HO-1, i.e., the downstream antioxidant proteins pertaining to Nrf2, while decreasing the damage of BBB and the degradation of tight junction proteins. To simulate the condition of hypoxia and ischemia in vitro, HBMEC cells received exposure to transient oxygen and glucose deprivation (OGD). The DCA treatment is capable of reducing the oxidative stress and blood-brain barrier of HBMEC cells after in vitro hypoxia and reperfusion (H/R). Furthermore, this study evidenced that HBMEC cells could exhibit higher susceptibility to H/R-induced oxidative stress after ML385 application, the specific inhibitor of Nrf2. Besides, the protection mediated by DCA disappeared after ML385 application. To sum up, as revealed from the mentioned results, DCA could exert the neuroprotective effect on oxidative stress and blood-brain barrier after brain I/R injury via PDK2-PDH-Nrf2 pathway activation. Accordingly, the PDK2-PDH-Nrf2 pathway may play a key role and provide a new pharmacology target in cerebral IS and I/R protection by DCA.
Collapse
Affiliation(s)
- Xiaoyong Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang 261021, China
| | - Shan Li
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yunchang Mo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Ruru Li
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Shaoyi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Anqi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Xuqing Ni
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Qinxue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| |
Collapse
|
23
|
Lajin B, Braeuer S, Borovička J, Goessler W. Is the water disinfection by-product dichloroacetic acid biosynthesized in the edible mushroom Russula nigricans? CHEMOSPHERE 2021; 281:130819. [PMID: 33991903 DOI: 10.1016/j.chemosphere.2021.130819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 06/12/2023]
Abstract
We report the first halogen speciation analysis study by high performance liquid chromatography coupled with inductively coupled plasma tandem mass spectrometry (HPLC-ICPMS/MS) in the fruiting bodies of various mushroom species. Non-targeted speciation analysis revealed the occurrence of dichloroacetic acid (DCAA) in the edible mushroom Russula nigricans. Multiple samples of this mushroom (n = 5) collected from different geographic non-industrial regions in two different countries confirmed the consistent presence of this species at a relatively narrow concentration range (23-37 mg kg-1), whereas no other chlorinated acetic acid (e.g. chloroacetic acid and trichloroacetic acid) was detected. Neither DCAA nor any other chlorinated acetic acid were detected in any of the other mushroom species investigated in the present study, including seven different mushroom species of the same genus Russula, even though all mushrooms were collected from the same non-industrial geographic regions. Together with the previously reported biological activities of DCAA, these findings collectively suggest biosynthesis of this compound as an explanation for its dominant presence in R. nigricans, and constitute the first example of the dominant natural occurrence of this compound over other chlorinated acetic acids in a living organism. This may warrant a change in our view of the occurrence of dichloroacetic acid in nature, where primarily considered as a pollutant arising from water disinfection.
Collapse
Affiliation(s)
- Bassam Lajin
- Institute of Chemistry, University of Graz, Universitaetsplatz 1, 8010, Graz, Austria.
| | - Simone Braeuer
- Institute of Chemistry, University of Graz, Universitaetsplatz 1, 8010, Graz, Austria; Atomic & Mass Spectrometry Research Unit, Department of Chemistry, Ghent University, Krijgslaan 281-S12, 9000, Ghent, Belgium
| | - Jan Borovička
- Nuclear Physics Institute of the Czech Academy of Sciences, Hlavní 130, 25068, Husinec-Řež, Czech Republic; Institute of Geology of the Czech Academy of Sciences, Rozvojová 269, 16500, Prague 6, Czech Republic
| | - Walter Goessler
- Institute of Chemistry, University of Graz, Universitaetsplatz 1, 8010, Graz, Austria
| |
Collapse
|
24
|
Mannaa FAE, Abdel-Wahhab KGED, Daoud EM, El Gendy AAR, Saber MM, Fadl NN. Effectiveness of low-power laser therapy in improvement of the peripheral neuropathy induced by xenobiotics in rats. Biochem Biophys Rep 2021; 27:101085. [PMID: 34381880 PMCID: PMC8334374 DOI: 10.1016/j.bbrep.2021.101085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Peripheral neuropathy (PN) is the damage and dysfunction of neurons of the peripheral nervous system. The present study was conducted to estimate the effectiveness of low-power laser therapy (LPLT) in the management of PN in a rats' model. METHODS PN was induced by giving dichloroacetate (DCA) (250 mg/kg/day) for up to 12 weeks. Four groups of rats were used: control group, PN group, PN group treated with gabapentin and PN group treated with LPLT. The study was conducted for 8 weeks. The management of PN was estimated by behavioral tests which included hot plate and Morris water maze tests. Blood biochemical analysis were carried out. RESULTS Using of hot plate test indicated thermal hypoalgesia and using Morris water maze test showed cognitive decline in PN rats. Treatment with LPLT or gabapentin improved both the pain sensations and deteriorated memory that occurred in the PN rats. Biochemical analysis showed that LPLT significantly decreased the elevated beta-endorphin level in PN rats, while gabapentin could not reduce it. Treatment PN rats with LPLT or gabapentin shifted the high levels of TNF-α, IL-1β and IL-10 cytokines back to their normal values. Serum nitric oxide and MDA significantly increased in the PN group together with significant reduction in the rGSH level, these values were significantly improved by LPLT application while this was not the case with gabapentin treatment. Furthermore, treatment with gabapentin or LPLT significantly reduced serum ALAT and ASAT activities which are otherwise increased in the PN group. S100B, PGE2, total cholesterol, triglycerides, LDL-cholesterol, HDL-cholesterol, urea and creatinine showed insignificant changes among all groups. CONCLUSIONS Our results showed that treatment with LPLT is more efficient than gabapentin in ameliorating the peripheral neuropathy induced by xenobiotics.
Collapse
Key Words
- ADP, adenosine diphosphate
- ATP, Adenosine triphosphate
- ATP, adenosine triphosphate
- DCA, Dichloroacetate
- Dichloroacetate
- Gabapentin
- IL-10, interleukin −10
- IL-1β, interleukin - 1β
- LPLT, Low power laser therapy
- Low-power laser therapy
- MCTs, monocarboxylate transporters
- MDA, malondialdehyde
- NAD+, Nicotinamide adenine dinucleotide
- NO, nitric oxide
- Neuropathy
- PDH, pyruvate dehydrogenase
- PGE2, prostaglandin E2
- PN, Peripheral neuropathy
- S100B, calcium binding protein B
- TCA, cycle tricarboxylic acid cycle or the Krebs cycle
- TNF-α, tumor necrosis factor- α
- rGSH, reduced glutathione
Collapse
Affiliation(s)
| | | | - Eitedal Mahmoud Daoud
- Complementary Medicine Department, National Research Centre, Dokki, Cairo, 12622, Egypt
| | | | - Maha Mohamed Saber
- Complementary Medicine Department, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Nevein Naim Fadl
- Medical Physiology Department, National Research Centre, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
25
|
Chen G, Jiang N, Villalobos Solis MI, Kara Murdoch F, Murdoch RW, Xie Y, Swift CM, Hettich RL, Löffler FE. Anaerobic Microbial Metabolism of Dichloroacetate. mBio 2021; 12:e00537-21. [PMID: 33906923 PMCID: PMC8092247 DOI: 10.1128/mbio.00537-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/23/2022] Open
Abstract
Dichloroacetate (DCA) commonly occurs in the environment due to natural production and anthropogenic releases, but its fate under anoxic conditions is uncertain. Mixed culture RM comprising "Candidatus Dichloromethanomonas elyunquensis" strain RM utilizes DCA as an energy source, and the transient formation of formate, H2, and carbon monoxide (CO) was observed during growth. Only about half of the DCA was recovered as acetate, suggesting a fermentative catabolic route rather than a reductive dechlorination pathway. Sequencing of 16S rRNA gene amplicons and 16S rRNA gene-targeted quantitative real-time PCR (qPCR) implicated "Candidatus Dichloromethanomonas elyunquensis" strain RM in DCA degradation. An (S)-2-haloacid dehalogenase (HAD) encoded on the genome of strain RM was heterologously expressed, and the purified HAD demonstrated the cofactor-independent stoichiometric conversion of DCA to glyoxylate at a rate of 90 ± 4.6 nkat mg-1 protein. Differential protein expression analysis identified enzymes catalyzing the conversion of DCA to acetyl coenzyme A (acetyl-CoA) via glyoxylate as well as enzymes of the Wood-Ljungdahl pathway. Glyoxylate carboligase, which catalyzes the condensation of two molecules of glyoxylate to form tartronate semialdehyde, was highly abundant in DCA-grown cells. The physiological, biochemical, and proteogenomic data demonstrate the involvement of an HAD and the Wood-Ljungdahl pathway in the anaerobic fermentation of DCA, which has implications for DCA turnover in natural and engineered environments, as well as the metabolism of the cancer drug DCA by gut microbiota.IMPORTANCE Dichloroacetate (DCA) is ubiquitous in the environment due to natural formation via biological and abiotic chlorination processes and the turnover of chlorinated organic materials (e.g., humic substances). Additional sources include DCA usage as a chemical feedstock and cancer drug and its unintentional formation during drinking water disinfection by chlorination. Despite the ubiquitous presence of DCA, its fate under anoxic conditions has remained obscure. We discovered an anaerobic bacterium capable of metabolizing DCA, identified the enzyme responsible for DCA dehalogenation, and elucidated a novel DCA fermentation pathway. The findings have implications for the turnover of DCA and the carbon and electron flow in electron acceptor-depleted environments and the human gastrointestinal tract.
Collapse
Affiliation(s)
- Gao Chen
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
- Department of Civil and Environmental Engineering, University of Tennessee, Knoxville, Tennessee, USA
| | - Nannan Jiang
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
- Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee, USA
- University of Tennessee and Oak Ridge National Laboratory (UT-ORNL) Joint Institute for Biological Sciences (JIBS), Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | | | - Fadime Kara Murdoch
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
- University of Tennessee and Oak Ridge National Laboratory (UT-ORNL) Joint Institute for Biological Sciences (JIBS), Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Robert Waller Murdoch
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
| | - Yongchao Xie
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
- Department of Civil and Environmental Engineering, University of Tennessee, Knoxville, Tennessee, USA
| | - Cynthia M Swift
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Robert L Hettich
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Frank E Löffler
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, Tennessee, USA
- Department of Civil and Environmental Engineering, University of Tennessee, Knoxville, Tennessee, USA
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
- Department of Biosystems Engineering & Soil Science, University of Tennessee, Knoxville, Tennessee, USA
- Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee, USA
- Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, USA
- University of Tennessee and Oak Ridge National Laboratory (UT-ORNL) Joint Institute for Biological Sciences (JIBS), Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| |
Collapse
|
26
|
Defective palmitoylation of transferrin receptor triggers iron overload in Friedreich ataxia fibroblasts. Blood 2021; 137:2090-2102. [PMID: 33529321 DOI: 10.1182/blood.2020006987] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Friedreich ataxia (FRDA) is a frequent autosomal recessive disease caused by a GAA repeat expansion in the FXN gene encoding frataxin, a mitochondrial protein involved in iron-sulfur cluster (ISC) biogenesis. Resulting frataxin deficiency affects ISC-containing proteins and causes iron to accumulate in the brain and heart of FRDA patients. Here we report on abnormal cellular iron homeostasis in FRDA fibroblasts inducing a massive iron overload in cytosol and mitochondria. We observe membrane transferrin receptor 1 (TfR1) accumulation, increased TfR1 endocytosis, and delayed Tf recycling, ascribing this to impaired TfR1 palmitoylation. Frataxin deficiency is shown to reduce coenzyme A (CoA) availability for TfR1 palmitoylation. Finally, we demonstrate that artesunate, CoA, and dichloroacetate improve TfR1 palmitoylation and decrease iron overload, paving the road for evidence-based therapeutic strategies at the actionable level of TfR1 palmitoylation in FRDA.
Collapse
|
27
|
Tinker RJ, Lim AZ, Stefanetti RJ, McFarland R. Current and Emerging Clinical Treatment in Mitochondrial Disease. Mol Diagn Ther 2021; 25:181-206. [PMID: 33646563 PMCID: PMC7919238 DOI: 10.1007/s40291-020-00510-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2020] [Indexed: 12/11/2022]
Abstract
Primary mitochondrial disease (PMD) is a group of complex genetic disorders that arise due to pathogenic variants in nuclear or mitochondrial genomes. Although PMD is one of the most prevalent inborn errors of metabolism, it often exhibits marked phenotypic variation and can therefore be difficult to recognise. Current treatment for PMD revolves around supportive and preventive approaches, with few disease-specific therapies available. However, over the last decade there has been considerable progress in our understanding of both the genetics and pathophysiology of PMD. This has resulted in the development of a plethora of new pharmacological and non-pharmacological therapies at varying stages of development. Many of these therapies are currently undergoing clinical trials. This review summarises the latest emerging therapies that may become mainstream treatment in the coming years. It is distinct from other recent reviews in the field by comprehensively addressing both pharmacological non-pharmacological therapy from both a bench and a bedside perspective. We highlight the current and developing therapeutic landscape in novel pharmacological treatment, dietary supplementation, exercise training, device use, mitochondrial donation, tissue replacement gene therapy, hypoxic therapy and mitochondrial base editing.
Collapse
Affiliation(s)
- Rory J Tinker
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Albert Z Lim
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Renae J Stefanetti
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- NHS Highly Specialised Service for Rare Mitochondrial Disorders for Adults and Children, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
28
|
Synthesis, biological evaluation and structure-activity relationship of novel dichloroacetophenones targeting pyruvate dehydrogenase kinases with potent anticancer activity. Eur J Med Chem 2021; 214:113225. [PMID: 33550182 DOI: 10.1016/j.ejmech.2021.113225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 01/25/2023]
Abstract
Pyruvate dehydrogenase kinases (PDKs) are promising therapeutic targets that have received increasing attentions in cancer metabolism. In this paper, we report the synthesis and biological evaluation of a series of novel dichloroacetophenones as potent PDKs inhibitors. Structure-activity relationship analysis enabled us to identify a potent compound 6u, which inhibited PDKs with an EC50 value of 0.09 μM, and reduced various cancer cells proliferation with IC50 values ranging from 1.1 to 3.8 μM, while show weak effect against non-cancerous L02 cell (IC50 > 10 μM). In the A375 xenograft model, 6u displayed an obvious antitumor activity at a dose of 5 mg/kg, but with no negative effect to the mice weight. Molecular docking suggested that 6u formed direct hydrogen bond interactions with Ser75 and Gln61 in PDK1, and meanwhile the aniline skeleton in 6u was sandwiched by the conserved hydrophobic residues Phe78 and Phe65, which contribute to the biochemical activity improvement. Moreover, 6u induced A375 cell apoptosis and cell arrest in G1 phase, and inhibited cancer cell migration. In addition, 6u altered glucose metabolic pathway in A375 cell by decreasing lactate formation and increasing ROS production and OCR consumption, which could serve as a potential modulator to reprogram the glycolysis pathway in cancer cell.
Collapse
|
29
|
Duraj T, García-Romero N, Carrión-Navarro J, Madurga R, Ortiz de Mendivil A, Prat-Acin R, Garcia-Cañamaque L, Ayuso-Sacido A. Beyond the Warburg Effect: Oxidative and Glycolytic Phenotypes Coexist within the Metabolic Heterogeneity of Glioblastoma. Cells 2021; 10:202. [PMID: 33498369 PMCID: PMC7922554 DOI: 10.3390/cells10020202] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor, with a median survival at diagnosis of 16-20 months. Metabolism represents a new attractive therapeutic target; however, due to high intratumoral heterogeneity, the application of metabolic drugs in GBM is challenging. We characterized the basal bioenergetic metabolism and antiproliferative potential of metformin (MF), dichloroacetate (DCA), sodium oxamate (SOD) and diazo-5-oxo-L-norleucine (DON) in three distinct glioma stem cells (GSCs) (GBM18, GBM27, GBM38), as well as U87MG. GBM27, a highly oxidative cell line, was the most resistant to all treatments, except DON. GBM18 and GBM38, Warburg-like GSCs, were sensitive to MF and DCA, respectively. Resistance to DON was not correlated with basal metabolic phenotypes. In combinatory experiments, radiomimetic bleomycin exhibited therapeutically relevant synergistic effects with MF, DCA and DON in GBM27 and DON in all other cell lines. MF and DCA shifted the metabolism of treated cells towards glycolysis or oxidation, respectively. DON consistently decreased total ATP production. Our study highlights the need for a better characterization of GBM from a metabolic perspective. Metabolic therapy should focus on both glycolytic and oxidative subpopulations of GSCs.
Collapse
Affiliation(s)
- Tomás Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, 28668 Madrid, Spain;
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Josefa Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Rodrigo Madurga
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | | | - Ricardo Prat-Acin
- Neurosurgery Department, Hospital Universitario La Fe, 46026 Valencia, Spain;
| | | | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.); (R.M.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| |
Collapse
|
30
|
Hu B, Boakye‐Yiadom KO, Yu W, Yuan Z, Ho W, Xu X, Zhang X. Nanomedicine Approaches for Advanced Diagnosis and Treatment of Atherosclerosis and Related Ischemic Diseases. Adv Healthc Mater 2020; 9:e2000336. [PMID: 32597562 DOI: 10.1002/adhm.202000336] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/30/2020] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVDs) remain one of the major causes of mortality worldwide. In response to this and other worldwide health epidemics, nanomedicine has emerged as a rapidly evolving discipline that involves the development of innovative nanomaterials and nanotechnologies and their applications in therapy and diagnosis. Nanomedicine presents unique advantages over conventional medicines due to the superior properties intrinsic to nanoscopic therapies. Once used mainly for cancer therapies, recently, tremendous progress has been made in nanomedicine that has led to an overall improvement in the treatment and diagnosis of CVDs. This review elucidates the pathophysiology and potential targets of atherosclerosis and associated ischemic diseases. It may be fruitful to pursue future work in the nanomedicine-mediated treatment of CVDs based on these targets. A comprehensive overview is then provided featuring the latest preclinical and clinical outcomes in cardiovascular imaging, biomarker detection, tissue engineering, and nanoscale delivery, with specific emphasis on nanoparticles, nanostructured scaffolds, and nanosensors. Finally, the challenges and opportunities regarding the future development and clinical translation of nanomedicine in related fields are discussed. Overall, this review aims to provide a deep and thorough understanding of the design, application, and future development of nanomedicine for atherosclerosis and related ischemic diseases.
Collapse
Affiliation(s)
- Bin Hu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Kofi Oti Boakye‐Yiadom
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Wei Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Zi‐Wei Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| |
Collapse
|
31
|
Kang J, Pagire HS, Kang D, Song YH, Lee IK, Lee KT, Park CJ, Ahn JH, Kim J. Structural basis for the inhibition of PDK2 by novel ATP- and lipoyl-binding site targeting compounds. Biochem Biophys Res Commun 2020; 527:778-784. [PMID: 32444142 DOI: 10.1016/j.bbrc.2020.04.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 11/19/2022]
Abstract
Pyruvate dehydrogenase kinase (PDK) controls the activity of pyruvate decarboxylase complex (PDC) by phosphorylating key serine residues on the E1 subunit, which leads to a decreased oxidative phosphorylation in mitochondria. Inhibition of PDK activity by natural/synthetic compounds has been shown to reverse the Warburg effect, a characteristic metabolism in cancer cells. PDK-PDC axis also has been associated with diabetes and heart disease. Therefore, regulation of PDK activity has been considered as a promising strategy to treat related diseases. Here we present the X-ray crystal structure of PDK2 complexed with a recently identified PDK4 inhibitor, compound 8c, which has been predicted to bind at the lipoyl-binding site and interrupt intermolecular interactions with the E2-E3bp subunits of PDC. The co-crystal structure confirmed the specific binding location of compound 8c and revealed the remote conformational change in the ATP-binding pocket. In addition, two novel 4,5-diarylisoxazole derivatives, GM10030 and GM67520, were synthesized and used for structural studies, which target the ATP-binding site of PDK2. These compounds bind to PDK2 with a sub-100nM affinity as determined by isothermal titration calorimetry experiments. Notably, the crystal structure of the PDK2-GM10030 complex displays unprecedented asymmetric conformation of human PDK2 dimer, especially in the ATP-lids and C-terminal tails.
Collapse
Affiliation(s)
- Jihoon Kang
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Haushabhau S Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Donguk Kang
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yo Han Song
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - In Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Kang Taek Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Chin-Ju Park
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jungwook Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
32
|
Lahiguera Á, Hyroššová P, Figueras A, Garzón D, Moreno R, Soto-Cerrato V, McNeish I, Serra V, Lazaro C, Barretina P, Brunet J, Menéndez J, Matias-Guiu X, Vidal A, Villanueva A, Taylor-Harding B, Tanaka H, Orsulic S, Junza A, Yanes O, Muñoz-Pinedo C, Palomero L, Pujana MÀ, Perales JC, Viñals F. Tumors defective in homologous recombination rely on oxidative metabolism: relevance to treatments with PARP inhibitors. EMBO Mol Med 2020; 12:e11217. [PMID: 32400970 PMCID: PMC7278557 DOI: 10.15252/emmm.201911217] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial metabolism and the generation of reactive oxygen species (ROS) contribute to the acquisition of DNA mutations and genomic instability in cancer. How genomic instability influences the metabolic capacity of cancer cells is nevertheless poorly understood. Here, we show that homologous recombination‐defective (HRD) cancers rely on oxidative metabolism to supply NAD+ and ATP for poly(ADP‐ribose) polymerase (PARP)‐dependent DNA repair mechanisms. Studies in breast and ovarian cancer HRD models depict a metabolic shift that includes enhanced expression of the oxidative phosphorylation (OXPHOS) pathway and its key components and a decline in the glycolytic Warburg phenotype. Hence, HRD cells are more sensitive to metformin and NAD+ concentration changes. On the other hand, shifting from an OXPHOS to a highly glycolytic metabolism interferes with the sensitivity to PARP inhibitors (PARPi) in these HRD cells. This feature is associated with a weak response to PARP inhibition in patient‐derived xenografts, emerging as a new mechanism to determine PARPi sensitivity. This study shows a mechanistic link between two major cancer hallmarks, which in turn suggests novel possibilities for specifically treating HRD cancers with OXPHOS inhibitors.
Collapse
Affiliation(s)
- Álvaro Lahiguera
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Petra Hyroššová
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| | - Agnès Figueras
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Diana Garzón
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Roger Moreno
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Vanessa Soto-Cerrato
- Departament de Patologia i Terapèutica Experimental, Universitat de Barcelona, Barcelona, Spain
| | - Iain McNeish
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Conxi Lazaro
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.,Hereditary Cancer Program, Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Pilar Barretina
- Medical Oncology Department, Institut Català d'Oncologia, IDIBGI, Girona, Spain
| | - Joan Brunet
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.,Hereditary Cancer Program, Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Hereditary Cancer Program, Institut Català d'Oncologia, IDIBGI, Girona, Spain.,Medical Sciences Department, School of Medicine, University of Girona, Girona, Spain
| | - Javier Menéndez
- Program against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Xavier Matias-Guiu
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.,Servei d'Anatomia Patològica, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - August Vidal
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Departament de Patologia i Terapèutica Experimental, Universitat de Barcelona, Barcelona, Spain.,Servei d'Anatomia Patològica, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain.,Xenopat, Carrer de la Feixa Llarga S/N, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alberto Villanueva
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Xenopat, Carrer de la Feixa Llarga S/N, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Hisashi Tanaka
- Womens Cancer Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sandra Orsulic
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Alexandra Junza
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, Tarragona, Spain.,Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Oscar Yanes
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, Tarragona, Spain.,Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luís Palomero
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Miquel Àngel Pujana
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - José Carlos Perales
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Viñals
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
33
|
Abstract
The mercapturic acid pathway is a major route for the biotransformation of xenobiotic and endobiotic electrophilic compounds and their metabolites. Mercapturic acids (N-acetyl-l-cysteine S-conjugates) are formed by the sequential action of the glutathione transferases, γ-glutamyltransferases, dipeptidases, and cysteine S-conjugate N-acetyltransferase to yield glutathione S-conjugates, l-cysteinylglycine S-conjugates, l-cysteine S-conjugates, and mercapturic acids; these metabolites constitute a "mercapturomic" profile. Aminoacylases catalyze the hydrolysis of mercapturic acids to form cysteine S-conjugates. Several renal transport systems facilitate the urinary elimination of mercapturic acids; urinary mercapturic acids may serve as biomarkers for exposure to chemicals. Although mercapturic acid formation and elimination is a detoxication reaction, l-cysteine S-conjugates may undergo bioactivation by cysteine S-conjugate β-lyase. Moreover, some l-cysteine S-conjugates, particularly l-cysteinyl-leukotrienes, exert significant pathophysiological effects. Finally, some enzymes of the mercapturic acid pathway are described as the so-called "moonlighting proteins," catalytic proteins that exert multiple biochemical or biophysical functions apart from catalysis.
Collapse
Affiliation(s)
- Patrick E Hanna
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
34
|
Prins KW, Thenappan T, Weir EK, Kalra R, Pritzker M, Archer SL. Repurposing Medications for Treatment of Pulmonary Arterial Hypertension: What's Old Is New Again. J Am Heart Assoc 2020; 8:e011343. [PMID: 30590974 PMCID: PMC6405714 DOI: 10.1161/jaha.118.011343] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kurt W Prins
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Thenappan Thenappan
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - E Kenneth Weir
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Rajat Kalra
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Marc Pritzker
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | | |
Collapse
|
35
|
Vengoji R, Ponnusamy MP, Rachagani S, Mahapatra S, Batra SK, Shonka N, Macha MA. Novel therapies hijack the blood-brain barrier to eradicate glioblastoma cancer stem cells. Carcinogenesis 2019; 40:2-14. [PMID: 30475990 DOI: 10.1093/carcin/bgy171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 10/12/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is amongst the most aggressive brain tumors with a dismal prognosis. Despite significant advances in the current multimodality therapy including surgery, postoperative radiotherapy (RT) and temozolomide (TMZ)-based concomitant and adjuvant chemotherapy (CT), tumor recurrence is nearly universal with poor patient outcomes. These limitations are in part due to poor drug penetration through the blood-brain barrier (BBB) and resistance to CT and RT by a small population of cancer cells recognized as tumor-initiating cells or cancer stem cells (CSCs). Though CT and RT kill the bulk of the tumor cells, they fail to affect CSCs, resulting in their enrichment and their development into more refractory tumors. Therefore, identifying the mechanisms of resistance and developing therapies that specifically target CSCs can improve response, prevent the development of refractory tumors and increase overall survival of GBM patients. Small molecule inhibitors that can breach the BBB and selectively target CSCs are emerging. In this review, we have summarized the recent advancements in understanding the GBM CSC-specific signaling pathways, the CSC-tumor microenvironment niche that contributes to CT and RT resistance and the use of novel combination therapies of small molecule inhibitors that may be used in conjunction with TMZ-based chemoradiation for effective management of GBM.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sidharth Mahapatra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
36
|
Twarock S, Reichert C, Bach K, Reiners O, Kretschmer I, Gorski DJ, Gorges K, Grandoch M, Fischer JW. Inhibition of the hyaluronan matrix enhances metabolic anticancer therapy by dichloroacetate in vitro and in vivo. Br J Pharmacol 2019; 176:4474-4490. [PMID: 31351004 PMCID: PMC6932941 DOI: 10.1111/bph.14808] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022] Open
Abstract
Background and Purpose Aerobic glycolysis is a unique feature of tumour cells that entails several advantages for cancer progression such as resistance to apoptosis. The low MW compound, dichloroacetate, is a pyruvate dehydrogenase kinase inhibitor, which restores oxidative phosphorylation and induces apoptosis in a variety of cancer entities. However, its therapeutic effectiveness is limited by resistance mechanisms. This study aimed to examine the role of the anti‐apoptotic hyaluronan (HA) matrix in this context and to identify a potential add‐on treatment option to overcome this limitation. Experimental Approach The metabolic connection between dichloroacetate treatment and HA matrix augmentation was analysed in vitro by quantitative PCR and affinity cytochemistry. Metabolic pathways were analysed using Seahorse, HPLC, fluorophore‐assisted carbohydrate electrophoresis, colourimetry, immunoblots, and immunochemistry. The effects of combining dichloroacetate with the HA synthesis inhibitor 4‐methylumbelliferone was evaluated in 2D and 3D cell cultures and in a nude mouse tumour xenograft regression model by immunoblot, immunochemistry, and FACS analysis. Key Results Mitochondrial reactivation induced by dichloroacetate metabolically activated HA synthesis by augmenting precursors as well as O‐GlcNAcylation. This process was blocked by 4‐methylumbelliferone, resulting in enhanced anti‐tumour efficacy in 2D and 3D cell culture and in a nude mouse tumour xenograft regression model. Conclusions and Implications The HA rich tumour micro‐environment represents a metabolic factor contributing to chemotherapy resistance. HA synthesis inhibition exhibited pronounced synergistic actions with dichloroacetate treatment on oesophageal tumour cell proliferation and survival in vitro and in vivo suggesting the combination of these two strategies is an effective anticancer therapy.
Collapse
Affiliation(s)
- Sören Twarock
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Christina Reichert
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Katharina Bach
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Oliver Reiners
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Inga Kretschmer
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Daniel J Gorski
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Katharina Gorges
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Maria Grandoch
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jens W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität, Düsseldorf, Germany
| |
Collapse
|
37
|
Chen TY, Hsieh YT, Huang JM, Liu CJ, Chuang LT, Huang PC, Kuo TY, Chia HY, Chou CY, Chang CW, Chen YF, Chen HM, Lo JF, Li WC. Determination of Pyruvate Metabolic Fates Modulates Head and Neck Tumorigenesis. Neoplasia 2019; 21:641-652. [PMID: 31100640 PMCID: PMC6522776 DOI: 10.1016/j.neo.2019.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/13/2019] [Accepted: 04/16/2019] [Indexed: 12/16/2022]
Abstract
Even with increasing evidence for roles of glycolytic enzymes in controlling cancerous characteristics, the best target of candidate metabolic enzymes for lessening malignancy remains under debate. Pyruvate is a main glycolytic metabolite that could be mainly converted into either lactate by Lactate Dehydrogenase A (LDHA) or acetyl-CoA by Pyruvate Dehydrogenase E1 component α subunit (PDHA1) catalytic complex. In tumor cells, accumulating lactate is produced whereas the conversion of pyruvate into mitochondrial acetyl-CoA is less active compared with their normal counterparts. This reciprocal molecular association makes pyruvate metabolism a potential choice of anti-cancer target. Cellular and molecular changes were herein assayed in Head and Neck Squamous Cell Carcinoma (HNSCC) cells in response to LDHA and PDHA1 loss in vitro, in vivo and in clinic. By using various human cancer databases and clinical samples, LDHA and PDHA1 levels exhibit reversed prognostic roles. In vitro analysis demonstrated that decreased cell growth and motility accompanied by an increased sensitivity to chemotherapeutic agents was found in cells with LDHA loss whereas PDHA1-silencing exhibited opposite phenotypes. At the molecular level, it was found that oncogenic Protein kinase B (PKB/Akt) and Extracellular signal-regulated kinase (ERK) singling pathways contribute to pyruvate metabolism mediated HNSCC cell growth. Furthermore, LDHA/PDHA1 changes in HNSCC cells resulted in a broad metabolic reprogramming while intracellular molecules including polyunsaturated fatty acids and nitrogen metabolism related metabolites underlie the malignant changes. Collectively, our findings reveal the significance of pyruvate metabolic fates in modulating HNSCC tumorigenesis and highlight the impact of metabolic plasticity in HNSCC cells.
Collapse
Key Words
- 4-nqo, 4-nitroquinoline 1-oxide
- 5-fu, 5-fluouracil
- abc, atp-binding cassette
- acacb, acetyl-coa carboxylase beta
- aldh, aldehyde dehydrogenase
- cddp, cisplatin
- dca, dicholoroacetate
- dlat, dihydrolipoamide s-acetyltransferase
- dld, dihydrolipoamide dehydrogenase
- don, 6-diazo-5-oxo-l-norlucine
- ecm, extracellular matrix
- egcg, epigallocatechin gallate
- emt, epithelial-mesenchymal transition
- eno, enolase
- erk, extracellular signal-regulated kinase
- fasn, fatty acid synthase
- hoscc, human oral squamous cell carcinoma
- gc-fid, gas chromatograph-flame ionization detector
- gls1, glutaminase 1
- gluts, glucose transporters
- glud1/2, glutamine dehydrogenase 1/2
- g3pdh, glyceraldehyde-3-phosphate dehydrogenase
- hnscc, head and neck squamous cell carcinoma
- ic50, half maximal inhibitory concentration
- inn, silibinin
- ldha, lactate dehydrogenase a
- lc–ms, liquid chromatography-mass spectrophotometry
- mufas, monounsaturated fatty acids
- mtt, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- nhok, normal human oral keratinocytes
- ocr, oxygen consumption rate
- os, overall survival
- oxphos, oxidative phosphorylation
- pdc, pyruvate dehydrogenase complex
- pdha1, pyruvate dehydrogenase e1 component α subunit
- pdk1, pyruvate dehydrogenase kinase 1
- pdp1, pyruvate dehydrogenase phosphatase 1
- pep, phosphoenolpyruvate
- pfk1, phosphofructokinase 1
- pgam1, phosphoglycerate mutase 1
- pkb/akt, protein kinase b
- pkm2, pyruvate kinase m2
- ppp, pentose phosphate pathway
- pufas, polyunsaturated fatty acids
- ros, reactive oxygen species
- scd1, stearoyl-coa desaturase 1
- sfas, saturated fatty acids
- shrna, short-hairpin rna
- srebf1/2, sterol regulatory element-binding transcription factor 1/2
- taxol, paclitaxel
- tca, tricarboxylic acid
- tcga, the cancer genomic atlas
Collapse
Affiliation(s)
- Tsai-Ying Chen
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Ta Hsieh
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Jian-Min Huang
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Chung-Ji Liu
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; Department of Oral and Maxillofacial Surgery, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Lu-Te Chuang
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Pei-Chun Huang
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Tz-Yu Kuo
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Yuan Chia
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Yi Chou
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Ching-Wen Chang
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Fen Chen
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Ming Chen
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Jeng-Fan Lo
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan; Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Wan-Chun Li
- Institute of Oral Biology, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan; Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
38
|
Zhang SL, Yang Z, Hu X, Tam KY. Dichloroacetophenones targeting at pyruvate dehydrogenase kinase 1 with improved selectivity and antiproliferative activity: Synthesis and structure-activity relationships. Bioorg Med Chem Lett 2018; 28:3441-3445. [DOI: 10.1016/j.bmcl.2018.09.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 01/25/2023]
|
39
|
Zhang SL, Yang Z, Hu X, Chakravarty H, Tam KY. Anticancer effects of some novel dichloroacetophenones through the inhibition of pyruvate dehydrogenase kinase 1. Eur J Pharm Sci 2018; 123:43-55. [DOI: 10.1016/j.ejps.2018.07.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/27/2022]
|
40
|
Lee EJ, Chung TW, Lee JH, Kim BS, Kim EY, Lee SO, Ha KT. Water-extracted branch of Cinnamomum cassia promotes lung cancer cell apoptosis by inhibiting pyruvate dehydrogenase kinase activity. J Pharmacol Sci 2018; 138:146-154. [DOI: 10.1016/j.jphs.2018.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/21/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022] Open
|
41
|
Zhang M, Cong Q, Zhang X, Zhang M, Lu Y, Xu C. Pyruvate dehydrogenase kinase 1 contributes to cisplatin resistance of ovarian cancer through EGFR activation. J Cell Physiol 2018; 234:6361-6370. [PMID: 30229902 DOI: 10.1002/jcp.27369] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Meng Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Qing Cong
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Xiao‐Yan Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai China
| | - Ming‐Xing Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Ying‐Ying Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Cong‐Jian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai China
| |
Collapse
|
42
|
The effect of dichloroacetate in mouse models of epilepsy. Epilepsy Res 2018; 145:77-81. [DOI: 10.1016/j.eplepsyres.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/17/2018] [Accepted: 06/09/2018] [Indexed: 01/23/2023]
|
43
|
Michelakis ED, Gurtu V, Webster L, Barnes G, Watson G, Howard L, Cupitt J, Paterson I, Thompson RB, Chow K, O'Regan DP, Zhao L, Wharton J, Kiely DG, Kinnaird A, Boukouris AE, White C, Nagendran J, Freed DH, Wort SJ, Gibbs JSR, Wilkins MR. Inhibition of pyruvate dehydrogenase kinase improves pulmonary arterial hypertension in genetically susceptible patients. Sci Transl Med 2018; 9:9/413/eaao4583. [PMID: 29070699 DOI: 10.1126/scitranslmed.aao4583] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/22/2017] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive vascular disease with a high mortality rate. It is characterized by an occlusive vascular remodeling due to a pro-proliferative and antiapoptotic environment in the wall of resistance pulmonary arteries (PAs). Proliferating cells exhibit a cancer-like metabolic switch where mitochondrial glucose oxidation is suppressed, whereas glycolysis is up-regulated as the major source of adenosine triphosphate production. This multifactorial mitochondrial suppression leads to inhibition of apoptosis and downstream signaling promoting proliferation. We report an increase in pyruvate dehydrogenase kinase (PDK), an inhibitor of the mitochondrial enzyme pyruvate dehydrogenase (PDH, the gatekeeping enzyme of glucose oxidation) in the PAs of human PAH compared to healthy lungs. Treatment of explanted human PAH lungs with the PDK inhibitor dichloroacetate (DCA) ex vivo activated PDH and increased mitochondrial respiration. In a 4-month, open-label study, DCA (3 to 6.25 mg/kg b.i.d.) administered to patients with idiopathic PAH (iPAH) already on approved iPAH therapies led to reduction in mean PA pressure and pulmonary vascular resistance and improvement in functional capacity, but with a range of individual responses. Lack of ex vivo and clinical response was associated with the presence of functional variants of SIRT3 and UCP2 that predict reduced protein function. Impaired function of these proteins causes PDK-independent mitochondrial suppression and pulmonary hypertension in mice. This first-in-human trial of a mitochondria-targeting drug in iPAH demonstrates that PDK is a druggable target and offers hemodynamic improvement in genetically susceptible patients, paving the way for novel precision medicine approaches in this disease.
Collapse
Affiliation(s)
| | - Vikram Gurtu
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Linda Webster
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Gareth Barnes
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Geoffrey Watson
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Luke Howard
- National Pulmonary Hypertension Service, Imperial College Healthcare National Health Service Trust, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - John Cupitt
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Ian Paterson
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Richard B Thompson
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Kelvin Chow
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Declan P O'Regan
- Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Lan Zhao
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - John Wharton
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| | - Adam Kinnaird
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | | | - Chris White
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Jayan Nagendran
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Darren H Freed
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Stephen J Wort
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| | - J Simon R Gibbs
- National Pulmonary Hypertension Service, Imperial College Healthcare National Health Service Trust, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Martin R Wilkins
- Department of Medicine, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
44
|
Hong DK, Kho AR, Choi BY, Lee SH, Jeong JH, Lee SH, Park KH, Park JB, Suh SW. Combined Treatment With Dichloroacetic Acid and Pyruvate Reduces Hippocampal Neuronal Death After Transient Cerebral Ischemia. Front Neurol 2018; 9:137. [PMID: 29593636 PMCID: PMC5857568 DOI: 10.3389/fneur.2018.00137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/26/2018] [Indexed: 12/28/2022] Open
Abstract
Transient cerebral ischemia (TCI) occurs when blood flow to the brain is ceased or dramatically reduced. TCI causes energy depletion and oxidative stress, which leads to neuronal death and cognitive impairment. Dichloroacetic acid (DCA) acts as an inhibitor of pyruvate dehydrogenase kinase (PDK). Additionally, DCA is known to increase mitochondrial pyruvate uptake and promotes glucose oxidation during glycolysis, thus enhancing pyruvate dehydrogenase (PDH) activity. In this study, we investigated whether the inhibition of PDK activity by DCA, which increases the rate of pyruvate conversion to adenosine triphosphate (ATP), prevents ischemia-induced neuronal death. We used a rat model of TCI, which was induced by common carotid artery occlusion and hypovolemia for 7 min while monitoring the electroencephalography for sustained isoelectric potential. Male Sprague-Dawley rats were given an intraperitoneal injection of DCA (100 mg/kg) with pyruvate (50 mg/kg) once per day for 2 days after insult. The vehicle, DCA only or pyruvate on rats was injected on the same schedule. Our study demonstrated that the combined administration of DCA with pyruvate significantly decreased neuronal death, oxidative stress, microglia activation when compared with DCA, or pyruvate injection alone. These findings suggest that the administration of DCA with pyruvate may enhance essential metabolic processes, which in turn promotes the regenerative capacity of the post-ischemic brain.
Collapse
Affiliation(s)
- Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - A Ra Kho
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Jeong Hyun Jeong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Sang Hwon Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Kyoung-Ha Park
- Division of Cardiovascular Diseases, Hallym University Sacred Heart Hospital, Anyang, South Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
45
|
Goodwin J, Choi H, Hsieh MH, Neugent ML, Ahn JM, Hayenga HN, Singh PK, Shackelford DB, Lee IK, Shulaev V, Dhar S, Takeda N, Kim JW. Targeting Hypoxia-Inducible Factor-1α/Pyruvate Dehydrogenase Kinase 1 Axis by Dichloroacetate Suppresses Bleomycin-induced Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2018; 58:216-231. [PMID: 28915065 DOI: 10.1165/rcmb.2016-0186oc] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia has long been implicated in the pathogenesis of fibrotic diseases. Aberrantly activated myofibroblasts are the primary pathological driver of fibrotic progression, yet how various microenvironmental influences, such as hypoxia, contribute to their sustained activation and differentiation is poorly understood. As a defining feature of hypoxia is its impact on cellular metabolism, we sought to investigate how hypoxia-induced metabolic reprogramming affects myofibroblast differentiation and fibrotic progression, and to test the preclinical efficacy of targeting glycolytic metabolism for the treatment of pulmonary fibrosis. Bleomycin-induced pulmonary fibrotic progression was evaluated in two independent, fibroblast-specific, promoter-driven, hypoxia-inducible factor (Hif) 1A knockout mouse models and in glycolytic inhibitor, dichloroacetate-treated mice. Genetic and pharmacological approaches were used to explicate the role of metabolic reprogramming in myofibroblast differentiation. Hypoxia significantly enhanced transforming growth factor-β-induced myofibroblast differentiation through HIF-1α, whereas overexpression of the critical HIF-1α-mediated glycolytic switch, pyruvate dehydrogenase kinase 1 (PDK1) was sufficient to activate glycolysis and potentiate myofibroblast differentiation, even in the absence of HIF-1α. Inhibition of the HIF-1α/PDK1 axis by genomic deletion of Hif1A or pharmacological inhibition of PDK1 significantly attenuated bleomycin-induced pulmonary fibrosis. Our findings suggest that HIF-1α/PDK1-mediated glycolytic reprogramming is a critical metabolic alteration that acts to promote myofibroblast differentiation and fibrotic progression, and demonstrate that targeting glycolytic metabolism may prove to be a potential therapeutic strategy for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | | | - Jung-Mo Ahn
- 2 Department of Chemistry and Biochemistry, and
| | - Heather N Hayenga
- 3 Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| | - Pankaj K Singh
- 4 Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - David B Shackelford
- 5 Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - In-Kyu Lee
- 6 Section of Endocrinology, Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Vladimir Shulaev
- 7 Department of Biological Sciences, College of Arts and Sciences, University of North Texas, Denton, Texas
| | - Shanta Dhar
- 8 Department of Biochemistry and Molecular Biology and.,9 Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida; and
| | - Norihiko Takeda
- 10 Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
46
|
Belkahla S, Haq Khan AU, Gitenay D, Alexia C, Gondeau C, Vo DN, Orecchioni S, Talarico G, Bertolini F, Cartron G, Hernandez J, Daujat-Chavanieu M, Allende-Vega N, Gonzalez MV. Changes in metabolism affect expression of ABC transporters through ERK5 and depending on p53 status. Oncotarget 2017; 9:1114-1129. [PMID: 29416681 PMCID: PMC5787424 DOI: 10.18632/oncotarget.23305] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 12/05/2017] [Indexed: 12/25/2022] Open
Abstract
Changes in metabolism require the efflux and influx of a diverse variety of metabolites. The ABC superfamily of transporters regulates the exchange of hundreds of substrates through the impermeable cell membrane. We show here that a metabolic switch to oxidative phosphorylation (OXPHOS), either by treating cells with dichloroacetate (DCA) or by changing the available substrates, reduced expression of ABCB1, ABCC1, ABCC5 and ABCG2 in wild-type p53-expressing cells. This metabolic change reduced histone changes associated to active promoters. Notably, DCA also inhibited expression of these genes in two animal models in vivo. In contrast, OXPHOS increased the expression of the same transporters in mutated (mut) or null p53-expressing cells. ABC transporters control the export of drugs from cancer cells and render tumors resistant to chemotherapy, playing an important role in multiple drug resistance (MDR). Wtp53 cells forced to perform OXPHOS showed impaired drug clearance. In contrast mutp53 cells increased drug clearance when performing OXPHOS. ABC transporter promoters contain binding sites for the transcription factors MEF2, NRF1 and NRF2 that are targets of the MAPK ERK5. OXPHOS induced expression of the MAPK ERK5. Decreasing ERK5 levels in wtp53 cells increased ABC expression whereas it inhibited expression in mutp53 cells. Our results showed that the ERK5/MEF2 pathway controlled ABC expression depending on p53 status.
Collapse
Affiliation(s)
- Sana Belkahla
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France
| | - Abrar Ul Haq Khan
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Delphine Gitenay
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Catherine Alexia
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Claire Gondeau
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France.,Département d'Hépato-gastroentérologie A, Hôpital Saint Eloi, CHU Montpellier, Montpellier, France
| | - Dang-Nghiem Vo
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France
| | - Stefania Orecchioni
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, Milan, Italy
| | - Giovanna Talarico
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, Milan, Italy
| | - Francesco Bertolini
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, Milan, Italy
| | - Guillaume Cartron
- Département d'Hématologie Clinique, CHU Montpellier, Université Montpellier I, Montpellier, France
| | - Javier Hernandez
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France
| | - Martine Daujat-Chavanieu
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Nerea Allende-Vega
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France.,These two authors share senior authorship
| | - Martin Villalba Gonzalez
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France.,These two authors share senior authorship
| |
Collapse
|
47
|
Phenyl butyrate inhibits pyruvate dehydrogenase kinase 1 and contributes to its anti-cancer effect. Eur J Pharm Sci 2017; 110:93-100. [DOI: 10.1016/j.ejps.2017.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/15/2022]
|
48
|
Han JE, Lim PW, Na CM, Choi YS, Lee JY, Kim Y, Park HW, Moon HE, Heo MS, Park HR, Kim DG, Paek SH. Inhibition of HIF1α and PDK Induces Cell Death of Glioblastoma Multiforme. Exp Neurobiol 2017; 26:295-306. [PMID: 29093638 PMCID: PMC5661062 DOI: 10.5607/en.2017.26.5.295] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/07/2017] [Accepted: 10/12/2017] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of brain tumors. GBMs, like other tumors, rely relatively less on mitochondrial oxidative phosphorylation (OXPHOS) and utilize more aerobic glycolysis, and this metabolic shift becomes augmented under hypoxia. In the present study, we investigated the physiological significance of altered glucose metabolism and hypoxic adaptation in the GBM cell line U251 and two newly established primary GBMs (GBM28 and GBM37). We found that these three GBMs exhibited differential growth rates under hypoxia compared to those under normoxia. Under normoxia, the basal expressions of HIF1α and the glycolysis-associated genes, PDK1, PDK3, and GLUT1, were relatively low in U251 and GBM28, while their basal expressions were high in GBM37. Under hypoxia, the expressions of these genes were enhanced further in all three GBMs. Treatment with dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK), induced cell death in GBM28 and GBM37 maintained under normoxia, whereas DCA effects disappeared under hypoxia, suggesting that hypoxic adaptation dominated DCA effects in these GBMs. In contrast, the inhibition of HIF1α with chrysin suppressed the expression of PDK1, PDK3, and GLUT1 and markedly promoted cell death of all GBMs under both normoxia and hypoxia. Interestingly, however, GBMs treated with chrysin under hypoxia still sustained higher viability than those under normoxia, and chrysin and DCA co-treatment was unable to eliminate this hypoxia-dependent resistance. Together, these results suggest that hypoxic adaptation is critical for maintaining viability of GBMs, and targeting hypoxic adaptation can be an important treatment option for GBMs.
Collapse
Affiliation(s)
- Jiwon Esther Han
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Pyung Won Lim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Chul Min Na
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - You Sik Choi
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Joo Young Lee
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yona Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyo Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Man Seung Heo
- Smart Healthcare Medical Device Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Hye Ran Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea.,Hypoxia Ischemia Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| |
Collapse
|
49
|
Wehmas LC, DeAngelo AB, Hester SD, Chorley BN, Carswell G, Olson GR, George MH, Carter JH, Eldridge SR, Fisher A, Vallanat B, Wood CE. Metabolic Disruption Early in Life is Associated With Latent Carcinogenic Activity of Dichloroacetic Acid in Mice. Toxicol Sci 2017; 159:354-365. [PMID: 28962523 PMCID: PMC6223632 DOI: 10.1093/toxsci/kfx146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Early-life environmental factors can influence later-life susceptibility to cancer. Recent evidence suggests that metabolic pathways may mediate this type of latency effect. Previously, we reported that short-term exposure to dichloroacetic acid (DCA) increased liver cancer in mice 84 weeks after exposure was stopped. Here, we evaluated time course dynamics for key events related to this effect. This study followed a stop-exposure design in which 28-day-old male B6C3F1 mice were given the following treatments in drinking water for up to 93 weeks: deionized water (dH2O, control); 3.5 g/l DCA continuously; or 3.5 g/l DCA for 4-52 weeks followed by dH2O. Effects were evaluated at eight interim time points. A short-term biomarker study was used to evaluate DCA effects at 6, 15, and 30 days. Liver tumor incidence was higher in all DCA treatment groups, including carcinomas in 82% of mice previously treated with DCA for only 4 weeks. Direct effects of DCA in the short-term study included decreased liver cell proliferation and marked mRNA changes related to mitochondrial dysfunction and altered cell metabolism. However, all observed short-term effects of DCA were ultimately reversible, and prior DCA treatment did not affect liver cell proliferation, apoptosis, necrosis, or DNA sequence variants with age. Key intermediate events resulting from transient DCA exposure do not fit classical cytotoxic, mitogenic, or genotoxic modes of action for carcinogenesis, suggesting a distinct mechanism associated with early-life metabolic disruption.
Collapse
Affiliation(s)
- Leah C. Wehmas
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Anthony B. DeAngelo
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Susan D. Hester
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Brian N. Chorley
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Gleta Carswell
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Greg R. Olson
- Toxicologic Pathology Associates, Jefferson, AK,
USA
| | - Michael H. George
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | | | | | - Anna Fisher
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Beena Vallanat
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Charles E. Wood
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| |
Collapse
|
50
|
Khan AUH, Allende-Vega N, Gitenay D, Gerbal-Chaloin S, Gondeau C, Vo DN, Belkahla S, Orecchioni S, Talarico G, Bertolini F, Bozic M, Valdivielso JM, Bejjani F, Jariel I, Lopez-Mejia IC, Fajas L, Lecellier CH, Hernandez J, Daujat M, Villalba M. The PDK1 Inhibitor Dichloroacetate Controls Cholesterol Homeostasis Through the ERK5/MEF2 Pathway. Sci Rep 2017; 7:10654. [PMID: 28878225 PMCID: PMC5587676 DOI: 10.1038/s41598-017-10339-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/08/2017] [Indexed: 12/17/2022] Open
Abstract
Controlling cholesterol levels is a major challenge in human health, since hypercholesterolemia can lead to serious cardiovascular disease. Drugs that target carbohydrate metabolism can also modify lipid metabolism and hence cholesterol plasma levels. In this sense, dichloroacetate (DCA), a pyruvate dehydrogenase kinase (PDK) inhibitor, augments usage of the glycolysis-produced pyruvate in the mitochondria increasing oxidative phosphorylation (OXPHOS). In several animal models, DCA decreases plasma cholesterol and triglycerides. Thus, DCA was used in the 70 s to treat diabetes mellitus, hyperlipoproteinemia and hypercholesterolemia with satisfactory results. However, the mechanism of action remained unknown and we describe it here. DCA increases LDLR mRNA and protein levels as well as LDL intake in several cell lines, primary human hepatocytes and two different mouse models. This effect is mediated by transcriptional activation as evidenced by H3 acetylation on lysine 27 on the LDLR promoter. DCA induces expression of the MAPK ERK5 that turns on the transcription factor MEF2. Inhibition of this ERK5/MEF2 pathway by genetic or pharmacological means decreases LDLR expression and LDL intake. In summary, our results indicate that DCA, by inducing OXPHOS, promotes ERK5/MEF2 activation leading to LDLR expression. The ERK5/MEF2 pathway offers an interesting pharmacological target for drug development.
Collapse
Affiliation(s)
- Abrar Ul Haq Khan
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France.,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France
| | - Nerea Allende-Vega
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France.,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France
| | - Delphine Gitenay
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France.,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France
| | - Sabine Gerbal-Chaloin
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France.,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France
| | - Claire Gondeau
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France.,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France.,Département d'Hépato-gastroentérologie A, Hôpital Saint Eloi, CHU, Montpellier, France
| | - Dang-Nghiem Vo
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France
| | - Sana Belkahla
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Giovanna Talarico
- Laboratory of Hematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Milica Bozic
- Vascular and Renal Translational Research Group. Institut de Recerca Biomedica de Lleida (IRBLLIDA), Lleida, Spain
| | - Jose M Valdivielso
- Vascular and Renal Translational Research Group. Institut de Recerca Biomedica de Lleida (IRBLLIDA), Lleida, Spain
| | | | | | | | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Javier Hernandez
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France
| | - Martine Daujat
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France.,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France
| | - Martin Villalba
- INSERM, U1183; Université de Montpellier, UFR Medecine, 80, av. Augustin Fliche, 34295, Montpellier Cedex 5, France. .,Institut de Médecine Régénératrice et Biothérapie (IRMB), CHU Montpellier, Montpellier, 34295, France.
| |
Collapse
|