1
|
Han J, Yuan Y, Zhang J, Hou Y, Xu H, Nie X, Zhao Z, Hou J. Regulatory effect of Wnt signaling on mitochondria in cancer: from mechanism to therapy. Apoptosis 2025:10.1007/s10495-025-02114-z. [PMID: 40257508 DOI: 10.1007/s10495-025-02114-z] [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] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Cancer is one of the most significant public health challenges in the new millennium, and complex mechanisms are at work to contribute to its pathogenesis and progression. The Wnt signaling pathways, which are crucial conserved cascades involved in embryological development and tissue homeostasis, and mitochondria, the intracellular powerhouses responsible for energy production, calcium and iron homeostasis, as well as mitochondrial apoptosis in eukaryotic cells, have their own mechanisms regulating these pathological processes. In the past decade, accumulating evidence has indicated that Wnt signaling pathways directly regulate mitochondrial biogenesis and function under physiological and pathological conditions. In this review, we systemically summarize the current understanding of how Wnt signaling pathways, particularly the canonical Wnt cascade, regulate mitochondrial fission, respiration, metabolism, and mitochondrial-dependent apoptosis in cancer. In addition, we discuss recent advancements in the research of anticancer agents and related pharmacological mechanisms targeting the signaling transduction of canonical Wnt pathway and/or mitochondrial function. We believe that the combined use of pharmaceuticals targeting Wnt signaling and/or mitochondria with conventional therapies, immunotherapy and targeted therapy based on accurate molecular pathological diagnosis will undoubtedly be the future mainstream direction of personalized cancer treatment, which could benefit more cancer patients.
Collapse
Affiliation(s)
- Jinping Han
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Yimeng Yuan
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Jianhua Zhang
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| | - Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Hongtao Xu
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China.
| | - Zhenhua Zhao
- Ma'anshan 86 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 243100, Ma'anshan, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| |
Collapse
|
2
|
Wang X, Rong C, Leng W, Niu P, He Z, Wang G, Qi X, Zhao D, Li J. Effect and mechanism of Dichloroacetate in the treatment of stroke and the resolution strategy for side effect. Eur J Med Res 2025; 30:148. [PMID: 40025562 PMCID: PMC11874805 DOI: 10.1186/s40001-025-02399-5] [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: 12/30/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Stroke is a serious disease that leads to high morbidity and mortality, and ischemic stroke accounts for more than 80% of strokes. At present, the only effective drug recombinant tissue plasminogen activator is limited by its indications, and its clinical application rate is not high. Therefore, it is urgent to develop effective new drugs according to the pathological mechanism. In the hypoxic state after ischemic stroke, anaerobic glycolysis has become the main way to provide energy to the brain. This process is essential for the maintenance of important brain functions and has important implications for recovery after stroke. However, acidosis caused by anaerobic glycolysis and lactic acid accumulation is an important pathological process after ischemic stroke. Dichloroacetate (DCA) is an orphan drug that has been used for decades to treat children with genetic mitochondrial diseases. Some studies have confirmed the role of DCA in stroke, but the conclusions are conflicting because some believe that DCA is not effective for ischemic stroke and may aggravate hemorrhagic stroke. This study reviews these studies and finds that DCA has a good effect on ischemic stroke. DCA can protect ischemic stroke by improving oxidative stress, reducing neuroinflammation, inhibiting apoptosis, protecting blood-brain barrier, and regulating metabolism. We also describe the differences in the outcomes of DCA in the treatment of ischemic stroke and the reasons why DCA aggravate hemorrhagic stroke. In addition, DCA, as a water disinfection byproduct, has been concerned about its toxicity. We describe the causes and solutions of peripheral neuropathy caused by DCA. In summary, this study analyzes the neuroprotective mechanism of DCA in ischemic stroke and the contradiction of the different research results, and discusses the causes and solutions of its adverse effects.
Collapse
Affiliation(s)
- Xu Wang
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chunshu Rong
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Wei Leng
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Ping Niu
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Ziqiao He
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Gaihua Wang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xin Qi
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Dexi Zhao
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
3
|
Mailloux RJ. The emerging importance of the α-keto acid dehydrogenase complexes in serving as intracellular and intercellular signaling platforms for the regulation of metabolism. Redox Biol 2024; 72:103155. [PMID: 38615490 PMCID: PMC11021975 DOI: 10.1016/j.redox.2024.103155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024] Open
Abstract
The α-keto acid dehydrogenase complex (KDHc) class of mitochondrial enzymes is composed of four members: pyruvate dehydrogenase (PDHc), α-ketoglutarate dehydrogenase (KGDHc), branched-chain keto acid dehydrogenase (BCKDHc), and 2-oxoadipate dehydrogenase (OADHc). These enzyme complexes occupy critical metabolic intersections that connect monosaccharide, amino acid, and fatty acid metabolism to Krebs cycle flux and oxidative phosphorylation (OxPhos). This feature also imbues KDHc enzymes with the heightened capacity to serve as platforms for propagation of intracellular and intercellular signaling. KDHc enzymes serve as a source and sink for mitochondrial hydrogen peroxide (mtH2O2), a vital second messenger used to trigger oxidative eustress pathways. Notably, deactivation of KDHc enzymes through reversible oxidation by mtH2O2 and other electrophiles modulates the availability of several Krebs cycle intermediates and related metabolites which serve as powerful intracellular and intercellular messengers. The KDHc enzymes also play important roles in the modulation of mitochondrial metabolism and epigenetic programming in the nucleus through the provision of various acyl-CoAs, which are used to acylate proteinaceous lysine residues. Intriguingly, nucleosomal control by acylation is also achieved through PDHc and KGDHc localization to the nuclear lumen. In this review, I discuss emerging concepts in the signaling roles fulfilled by the KDHc complexes. I highlight their vital function in serving as mitochondrial redox sensors and how this function can be used by cells to regulate the availability of critical metabolites required in cell signaling. Coupled with this, I describe in detail how defects in KDHc function can cause disease states through the disruption of cell redox homeodynamics and the deregulation of metabolic signaling. Finally, I propose that the intracellular and intercellular signaling functions of the KDHc enzymes are controlled through the reversible redox modification of the vicinal lipoic acid thiols in the E2 subunit of the complexes.
Collapse
Affiliation(s)
- Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
4
|
Wen L, Liu Z, Zhou L, Liu Z, Li Q, Geng B, Xia Y. Bone and Extracellular Signal-Related Kinase 5 (ERK5). Biomolecules 2024; 14:556. [PMID: 38785963 PMCID: PMC11117709 DOI: 10.3390/biom14050556] [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: 03/26/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Bones are vital for anchoring muscles, tendons, and ligaments, serving as a fundamental element of the human skeletal structure. However, our understanding of bone development mechanisms and the maintenance of bone homeostasis is still limited. Extracellular signal-related kinase 5 (ERK5), a recently identified member of the mitogen-activated protein kinase (MAPK) family, plays a critical role in the pathogenesis and progression of various diseases, especially neoplasms. Recent studies have highlighted ERK5's significant role in both bone development and bone-associated pathologies. This review offers a detailed examination of the latest research on ERK5 in different tissues and diseases, with a particular focus on its implications for bone health. It also examines therapeutic strategies and future research avenues targeting ERK5.
Collapse
Affiliation(s)
- Lei Wen
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Zirui Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Libo Zhou
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Zhongcheng Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Qingda Li
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| |
Collapse
|
5
|
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
|
6
|
Chalifoux O, Faerman B, Mailloux RJ. Mitochondrial hydrogen peroxide production by pyruvate dehydrogenase and α-ketoglutarate dehydrogenase in oxidative eustress and oxidative distress. J Biol Chem 2023; 299:105399. [PMID: 37898400 PMCID: PMC10692731 DOI: 10.1016/j.jbc.2023.105399] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023] Open
Abstract
Pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase (KGDH) are vital entry points for monosaccharides and amino acids into the Krebs cycle and thus integral for mitochondrial bioenergetics. Both complexes produce mitochondrial hydrogen peroxide (mH2O2) and are deactivated by electrophiles. Here, we provide an update on the role of PDH and KGDH in mitochondrial redox balance and their function in facilitating metabolic reprogramming for the propagation of oxidative eustress signals in hepatocytes and how defects in these pathways can cause liver diseases. PDH and KGDH are known to account for ∼45% of the total mH2O2 formed by mitochondria and display rates of production several-fold higher than the canonical source complex I. This mH2O2 can also be formed by reverse electron transfer (RET) in vivo, which has been linked to metabolic dysfunctions that occur in pathogenesis. However, the controlled emission of mH2O2 from PDH and KGDH has been proposed to be fundamental for oxidative eustress signal propagation in several cellular contexts. Modification of PDH and KGDH with protein S-glutathionylation (PSSG) and S-nitrosylation (PSNO) adducts serves as a feedback inhibitor for mH2O2 production in response to glutathione (GSH) pool oxidation. PSSG and PSNO adduct formation also reprogram the Krebs cycle to generate metabolites vital for interorganelle and intercellular signaling. Defects in the redox modification of PDH and KGDH cause the over generation of mH2O2, resulting in oxidative distress and metabolic dysfunction-associated fatty liver disease (MAFLD). In aggregate, PDH and KGDH are essential platforms for emitting and receiving oxidative eustress signals.
Collapse
Affiliation(s)
- Olivia Chalifoux
- Faculty of Agricultural and Environmental Sciences, The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ben Faerman
- Faculty of Agricultural and Environmental Sciences, The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- Faculty of Agricultural and Environmental Sciences, The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
7
|
Sohrabi A, Lefebvre AEYT, Harrison MJ, Condro MC, Sanazzaro TM, Safarians G, Solomon I, Bastola S, Kordbacheh S, Toh N, Kornblum HI, Digman MA, Seidlits SK. Microenvironmental stiffness induces metabolic reprogramming in glioblastoma. Cell Rep 2023; 42:113175. [PMID: 37756163 PMCID: PMC10842372 DOI: 10.1016/j.celrep.2023.113175] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The mechanical properties of solid tumors influence tumor cell phenotype and the ability to invade surrounding tissues. Using bioengineered scaffolds to provide a matrix microenvironment for patient-derived glioblastoma (GBM) spheroids, this study demonstrates that a soft, brain-like matrix induces GBM cells to shift to a glycolysis-weighted metabolic state, which supports invasive behavior. We first show that orthotopic murine GBM tumors are stiffer than peritumoral brain tissues, but tumor stiffness is heterogeneous where tumor edges are softer than the tumor core. We then developed 3D scaffolds with μ-compressive moduli resembling either stiffer tumor core or softer peritumoral brain tissue. We demonstrate that the softer matrix microenvironment induces a shift in GBM cell metabolism toward glycolysis, which manifests in lower proliferation rate and increased migration activities. Finally, we show that these mechanical cues are transduced from the matrix via CD44 and integrin receptors to induce metabolic and phenotypic changes in cancer cells.
Collapse
Affiliation(s)
- Alireza Sohrabi
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Austin E Y T Lefebvre
- Department of Biomedical Engineering, University of California at Irvine, Irvine, CA 92697, USA
| | - Mollie J Harrison
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Michael C Condro
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Talia M Sanazzaro
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Gevick Safarians
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Itay Solomon
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Soniya Bastola
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shadi Kordbacheh
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nadia Toh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Harley I Kornblum
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michelle A Digman
- Department of Biomedical Engineering, University of California at Irvine, Irvine, CA 92697, USA
| | - Stephanie K Seidlits
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
8
|
Jin AH, Qian YF, Ren J, Wang JG, Qiao F, Zhang ML, Du ZY, Luo Y. PDK inhibition promotes glucose utilization, reduces hepatic lipid deposition, and improves oxidative stress in largemouth bass (Micropterus salmoides) by increasing pyruvate oxidative phosphorylation. FISH & SHELLFISH IMMUNOLOGY 2023; 140:108969. [PMID: 37488039 DOI: 10.1016/j.fsi.2023.108969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
In omnivorous fish, the pyruvate dehydrogenase kinases (PDKs)-pyruvate dehydrogenase E1α subunit (PDHE1α) axis is essential in the regulation of carbohydrate oxidative catabolism. Among the existing research, the role of the PDKs-PDHE1α axis in carnivorous fish with poor glucose utilization is unclear. In the present study, we determined the effects of PDK inhibition on the liver glycolipid metabolism of largemouth bass (Micropterus salmoides). DCA is a PDK-specific inhibitor that inhibits PDK by binding the allosteric sites. A total of 160 juvenile largemouth bass were randomly divided into two groups, with four replicates of 20 fish each, fed a control diet and a control diet supplemented with dichloroacetate (DCA) for 8 weeks. The present results showed that DCA supplementation significantly decreased the hepatosomatic index, triglycerides in liver and serum, and total liver lipids of largemouth bass compared with the control group. In addition, compared with the control group, DCA treatment significantly down-regulated gene expression associated with lipogenesis. Furthermore, DCA supplementation significantly decreased the mRNA expression of pdk3a and increased PDHE1α activity. In addition, DCA supplementation improved glucose oxidative catabolism and pyruvate oxidative phosphorylation (OXPHOS) in the liver, as evidenced by low pyruvate content in the liver and up-regulated expressions of glycolysis-related and TCA cycle/OXPHOS-related genes. Moreover, DCA consumption decreased hepatic malondialdehyde (MDA) content, enhanced the activities of superoxide dismutase (SOD), and increased transforming growth factor beta (tgf-β), glutathione S-transferase (gst), and superoxide dismutase 1 (sod1) gene expression compared with the control diet. This study demonstrated that inhibition of PDKs by DCA promoted glucose utilization, reduced hepatic lipid deposition, and improved oxidative stress in largemouth bass by increasing pyruvate OXPHOS. Our findings contribute to the understanding of the underlying mechanism of the PDKs-PDHE1α axis in glucose metabolism and improve the utilization of dietary carbohydrates in farmed carnivorous fish.
Collapse
Affiliation(s)
- An-Hui Jin
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi-Fan Qian
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiong Ren
- HANOVE Research Center, Wuxi, PR China
| | - Jin-Gang Wang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Fang Qiao
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yuan Luo
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
9
|
Stancioiu F, Bogdan R, Dumitrescu R. Neuron-Specific Enolase (NSE) as a Biomarker for Autistic Spectrum Disease (ASD). Life (Basel) 2023; 13:1736. [PMID: 37629593 PMCID: PMC10455327 DOI: 10.3390/life13081736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Autistic spectrum disease (ASD) is an increasingly common diagnosis nowadays with a prevalence of 1-2% in most countries. Its complex causality-a combination of genetic, immune, metabolic, and environmental factors-is translated into pleiomorphic developmental disorders of various severity, which have two main aspects in common: repetitive, restrictive behaviors and difficulties in social interaction varying from awkward habits and verbalization to a complete lack of interest for the outside world. The wide variety of ASD causes also makes it very difficult to find a common denominator-a disease biomarker and medication-and currently, there is no commonly used diagnostic and therapeutic strategy besides clinical evaluation and psychotherapy. In the CORDUS clinical study, we have administered autologous cord blood to ASD kids who had little or no improvement after other treatments and searched for a biomarker which could help predict the degree of improvement in each patient. We have found that the neuron-specific enolase (NSE) was elevated above the normal clinical range (less than 16.3 ng/mL) in the vast majority of ASD kids tested in our study (40 of 41, or 97.5%). This finding opens up a new direction for diagnostic confirmation, dynamic evaluation, and therapeutic intervention for ASD kids.
Collapse
Affiliation(s)
| | - Raluca Bogdan
- Medicover Hospital Bucharest, 013982 Bucharest, Romania
| | | |
Collapse
|
10
|
Forteza MJ, Berg M, Edsfeldt A, Sun J, Baumgartner R, Kareinen I, Casagrande FB, Hedin U, Zhang S, Vuckovic I, Dzeja PP, Polyzos KA, Gisterå A, Trauelsen M, Schwartz TW, Dib L, Herrmann J, Monaco C, Matic L, Gonçalves I, Ketelhuth DFJ. Pyruvate dehydrogenase kinase regulates vascular inflammation in atherosclerosis and increases cardiovascular risk. Cardiovasc Res 2023; 119:1524-1536. [PMID: 36866436 PMCID: PMC10318388 DOI: 10.1093/cvr/cvad038] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/11/2023] [Accepted: 02/01/2023] [Indexed: 03/04/2023] Open
Abstract
AIMS Recent studies have revealed a close connection between cellular metabolism and the chronic inflammatory process of atherosclerosis. While the link between systemic metabolism and atherosclerosis is well established, the implications of altered metabolism in the artery wall are less understood. Pyruvate dehydrogenase kinase (PDK)-dependent inhibition of pyruvate dehydrogenase (PDH) has been identified as a major metabolic step regulating inflammation. Whether the PDK/PDH axis plays a role in vascular inflammation and atherosclerotic cardiovascular disease remains unclear. METHODS AND RESULTS Gene profiling of human atherosclerotic plaques revealed a strong correlation between PDK1 and PDK4 transcript levels and the expression of pro-inflammatory and destabilizing genes. Remarkably, the PDK1 and PDK4 expression correlated with a more vulnerable plaque phenotype, and PDK1 expression was found to predict future major adverse cardiovascular events. Using the small-molecule PDK inhibitor dichloroacetate (DCA) that restores arterial PDH activity, we demonstrated that the PDK/PDH axis is a major immunometabolic pathway, regulating immune cell polarization, plaque development, and fibrous cap formation in Apoe-/- mice. Surprisingly, we discovered that DCA regulates succinate release and mitigates its GPR91-dependent signals promoting NLRP3 inflammasome activation and IL-1β secretion by macrophages in the plaque. CONCLUSIONS We have demonstrated for the first time that the PDK/PDH axis is associated with vascular inflammation in humans and particularly that the PDK1 isozyme is associated with more severe disease and could predict secondary cardiovascular events. Moreover, we demonstrate that targeting the PDK/PDH axis with DCA skews the immune system, inhibits vascular inflammation and atherogenesis, and promotes plaque stability features in Apoe-/- mice. These results point toward a promising treatment to combat atherosclerosis.
Collapse
Affiliation(s)
- Maria J Forteza
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Martin Berg
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Andreas Edsfeldt
- Cardiovascular Research Translational Studies, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, 20 502, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Carl-Bertil Laurells gata 9, 21 428, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Jan Waldenströms gata 35, 20 502, Malmö, Sweden
| | - Jangming Sun
- Cardiovascular Research Translational Studies, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, 20 502, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Carl-Bertil Laurells gata 9, 21 428, Malmö, Sweden
| | - Roland Baumgartner
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Ilona Kareinen
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Felipe Beccaria Casagrande
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Song Zhang
- Mayo Clinic Metabolomics Core, Mayo Clinic, 200, First St. SW Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200, First St. SW Rochester, MN 55905, USA
| | - Ivan Vuckovic
- Mayo Clinic Metabolomics Core, Mayo Clinic, 200, First St. SW Rochester, MN 55905, USA
| | - Petras P Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, 200, First St. SW Rochester, MN 55905, USA
| | - Konstantinos A Polyzos
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Anton Gisterå
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Mette Trauelsen
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Blegdamsvej 3A, 2200, Copenhagen, Denmark
| | - Thue W Schwartz
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Blegdamsvej 3A, 2200, Copenhagen, Denmark
| | - Lea Dib
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7FY, UK
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, 200, First St. SW Rochester, MN 55905, USA
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7FY, UK
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
| | - Isabel Gonçalves
- Cardiovascular Research Translational Studies, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, 20 502, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Carl-Bertil Laurells gata 9, 21 428, Malmö, Sweden
| | - Daniel F J Ketelhuth
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Instutet,BioClinicum, Solnavägen 30, Solna, 17 164, Stockholm, Sweden
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløws vej 21, 5000 Odense, Denmark
| |
Collapse
|
11
|
Leung CLK, Karunakaran S, Atser MG, Innala L, Hu X, Viau V, Johnson JD, Clee SM. Analysis of a genetic region affecting mouse body weight. Physiol Genomics 2023; 55:132-146. [PMID: 36717164 PMCID: PMC10042608 DOI: 10.1152/physiolgenomics.00137.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Genetic factors affect an individual's risk of developing obesity, but in most cases each genetic variant has a small effect. Discovery of genes that regulate obesity may provide clues about its underlying biological processes and point to new ways the disease can be treated. Preclinical animal models facilitate genetic discovery in obesity because environmental factors can be better controlled compared with the human population. We studied inbred mouse strains to identify novel genes affecting obesity and glucose metabolism. BTBR T+ Itpr3tf/J (BTBR) mice are fatter and more glucose intolerant than C57BL/6J (B6) mice. Prior genetic studies of these strains identified an obesity locus on chromosome 2. Using congenic mice, we found that obesity was affected by a ∼316 kb region, with only two known genes, pyruvate dehydrogenase kinase 1 (Pdk1) and integrin α 6 (Itga6). Both genes had mutations affecting their amino acid sequence and reducing mRNA levels. Both genes have known functions that could modulate obesity, lipid metabolism, insulin secretion, and/or glucose homeostasis. We hypothesized that genetic variation in or near Pdk1 or Itga6 causing reduced Pdk1 and Itga6 expression would promote obesity and impaired glucose tolerance. We used knockout mice lacking Pdk1 or Itga6 fed an obesigenic diet to test this hypothesis. Under the conditions we studied, we were unable to detect an individual contribution of either Pdk1 or Itga6 to body weight. During our studies, with conditions outside our control, we were unable to reproduce some of our previous body weight data. However, we identified a previously unknown role for Pdk1 in cardiac cholesterol metabolism providing the basis for future investigations. The studies described in this paper highlight the importance and the challenge using physiological outcomes to study obesity genes in mice.
Collapse
Affiliation(s)
- Connie L K Leung
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Subashini Karunakaran
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael G Atser
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leyla Innala
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiaoke Hu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Victor Viau
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Susanne M Clee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Proteogenomics of diffuse gliomas reveal molecular subtypes associated with specific therapeutic targets and immune-evasion mechanisms. Nat Commun 2023; 14:505. [PMID: 36720864 PMCID: PMC9889805 DOI: 10.1038/s41467-023-36005-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/12/2023] [Indexed: 02/02/2023] Open
Abstract
Diffuse gliomas are devastating brain tumors. Here, we perform a proteogenomic profiling of 213 retrospectively collected glioma tumors. Proteogenomic analysis reveals the downstream biological events leading by EGFR-, IDH1-, TP53-mutations. The comparative analysis illustrates the distinctive features of GBMs and LGGs, indicating CDK2 inhibitor might serve as a promising drug target for GBMs. Further proteogenomic integrative analysis combined with functional experiments highlight the cis-effect of EGFR alterations might lead to glioma tumor cell proliferation through ERK5 medicates nucleotide synthesis process. Proteome-based stratification of gliomas defines 3 proteomic subgroups (S-Ne, S-Pf, S-Im), which could serve as a complement to WHO subtypes, and would provide the essential framework for the utilization of specific targeted therapies for particular glioma subtypes. Immune clustering identifies three immune subtypes with distinctive immune cell types. Further analysis reveals higher EGFR alteration frequencies accounts for elevation of immune check point protein: PD-L1 and CD70 in T-cell infiltrated tumors.
Collapse
|
13
|
Gómez-Mellado VE, Chang JC, Ho-Mok KS, Bernardino Morcillo C, Kersten RHJ, Oude Elferink RPJ, Verhoeven AJ, Paulusma CC. ATP8B1 Deficiency Results in Elevated Mitochondrial Phosphatidylethanolamine Levels and Increased Mitochondrial Oxidative Phosphorylation in Human Hepatoma Cells. Int J Mol Sci 2022; 23:ijms232012344. [PMID: 36293199 PMCID: PMC9604224 DOI: 10.3390/ijms232012344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/20/2022] Open
Abstract
ATP8B1 is a phospholipid flippase that is deficient in patients with progressive familial intrahepatic cholestasis type 1 (PFIC1). PFIC1 patients suffer from severe liver disease but also present with dyslipidemia, including low plasma cholesterol, of yet unknown etiology. Here we show that ATP8B1 knockdown in HepG2 cells leads to a strong increase in the mitochondrial oxidative phosphorylation (OXPHOS) without a change in glycolysis. The enhanced OXPHOS coincides with elevated low-density lipoprotein receptor protein and increased mitochondrial fragmentation and phosphatidylethanolamine levels. Furthermore, expression of phosphatidylethanolamine N-methyltransferase, an enzyme that catalyzes the conversion of mitochondrial-derived phosphatidylethanolamine to phosphatidylcholine, was reduced in ATP8B1 knockdown cells. We conclude that ATP8B1 deficiency results in elevated mitochondrial PE levels that stimulate mitochondrial OXPHOS. The increased OXPHOS leads to elevated LDLR levels, which provides a possible explanation for the reduced plasma cholesterol levels in PFIC1 disease.
Collapse
Affiliation(s)
- Valentina E. Gómez-Mellado
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Jung-Chin Chang
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Kam S. Ho-Mok
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Carmen Bernardino Morcillo
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
| | - Remco H. J. Kersten
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Ronald P. J. Oude Elferink
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Arthur J. Verhoeven
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Coen C. Paulusma
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
14
|
Glucose Starvation or Pyruvate Dehydrogenase Activation Induce a Broad, ERK5-Mediated, Metabolic Remodeling Leading to Fatty Acid Oxidation. Cells 2022; 11:cells11091392. [PMID: 35563698 PMCID: PMC9104157 DOI: 10.3390/cells11091392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/05/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cells have metabolic flexibility that allows them to adapt to changes in substrate availability. Two highly relevant metabolites are glucose and fatty acids (FA), and hence, glycolysis and fatty acid oxidation (FAO) are key metabolic pathways leading to energy production. Both pathways affect each other, and in the absence of one substrate, metabolic flexibility allows cells to maintain sufficient energy production. Here, we show that glucose starvation or sustained pyruvate dehydrogenase (PDH) activation by dichloroacetate (DCA) induce large genetic remodeling to propel FAO. The extracellular signal-regulated kinase 5 (ERK5) is a key effector of this multistep metabolic remodeling. First, there is an increase in the lipid transport by expression of low-density lipoprotein receptor-related proteins (LRP), e.g., CD36, LRP1 and others. Second, an increase in the expression of members of the acyl-CoA synthetase long-chain (ACSL) family activates FA. Finally, the expression of the enzymes that catalyze the initial step in each cycle of FAO, i.e., the acyl-CoA dehydrogenases (ACADs), is induced. All of these pathways lead to enhanced cellular FAO. In summary, we show here that different families of enzymes, which are essential to perform FAO, are regulated by the signaling pathway, i.e., MEK5/ERK5, which transduces changes from the environment to genetic adaptations.
Collapse
|
15
|
Tiersma JF, Evers B, Bakker BM, Jalving M, de Jong S. Pyruvate Dehydrogenase Kinase Inhibition by Dichloroacetate in Melanoma Cells Unveils Metabolic Vulnerabilities. Int J Mol Sci 2022; 23:ijms23073745. [PMID: 35409102 PMCID: PMC8999016 DOI: 10.3390/ijms23073745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 12/03/2022] Open
Abstract
Melanoma is characterized by high glucose uptake, partially mediated through elevated pyruvate dehydrogenase kinase (PDK), making PDK a potential treatment target in melanoma. We aimed to reduce glucose uptake in melanoma cell lines through PDK inhibitors dichloroacetate (DCA) and AZD7545 and through PDK knockdown, to inhibit cell growth and potentially unveil metabolic co-vulnerabilities resulting from PDK inhibition. MeWo cells were most sensitive to DCA, while SK-MEL-2 was the least sensitive, with IC50 values ranging from 13.3 to 27.0 mM. DCA strongly reduced PDH phosphorylation and increased the oxygen consumption rate:extracellular acidification rate (OCR:ECAR) ratio up to 6-fold. Knockdown of single PDK isoforms had similar effects on PDH phosphorylation and OCR:ECAR ratio as DCA but did not influence sensitivity to DCA. Growth inhibition by DCA was synergistic with the glutaminase inhibitor CB-839 (2- to 5-fold sensitization) and with diclofenac, known to inhibit monocarboxylate transporters (MCTs) (3- to 8-fold sensitization). CB-839 did not affect the OCR:ECAR response to DCA, whereas diclofenac strongly inhibited ECAR and further increased the OCR:ECAR ratio. We conclude that in melanoma cell lines, DCA reduces proliferation through reprogramming of cellular metabolism and synergizes with other metabolically targeted drugs.
Collapse
Affiliation(s)
- Jiske F. Tiersma
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Bernard Evers
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (B.E.); (B.M.B.)
| | - Barbara M. Bakker
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (B.E.); (B.M.B.)
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: (M.J.); (S.d.J.); Tel.: +31-50-3615692 (M.J.); +31-50-3612964 (S.d.J.)
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: (M.J.); (S.d.J.); Tel.: +31-50-3615692 (M.J.); +31-50-3612964 (S.d.J.)
| |
Collapse
|
16
|
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: 4.0] [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
|
17
|
Clinical Significance and Regulation of ERK5 Expression and Function in Cancer. Cancers (Basel) 2022; 14:cancers14020348. [PMID: 35053510 PMCID: PMC8773716 DOI: 10.3390/cancers14020348] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/08/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is a unique kinase among MAPKs family members, given its large structure characterized by the presence of a unique C-terminal domain. Despite increasing data demonstrating the relevance of the ERK5 pathway in the growth, survival, and differentiation of normal cells, ERK5 has recently attracted the attention of several research groups given its relevance in inflammatory disorders and cancer. Accumulating evidence reported its role in tumor initiation and progression. In this review, we explore the gene expression profile of ERK5 among cancers correlated with its clinical impact, as well as the prognostic value of ERK5 and pERK5 expression levels in tumors. We also summarize the importance of ERK5 in the maintenance of a cancer stem-like phenotype and explore the major known contributions of ERK5 in the tumor-associated microenvironment. Moreover, although several questions are still open concerning ERK5 molecular regulation, different ERK5 isoforms derived from the alternative splicing process are also described, highlighting the potential clinical relevance of targeting ERK5 pathways.
Collapse
|
18
|
Yılmaz A, Tekin SB, Bilici M, Yılmaz H. The Significance of Controlling Nutritional Status (CONUT) Score as a Novel Prognostic Parameter in Small Cell Lung Cancer. Lung 2020; 198:695-704. [PMID: 32424800 DOI: 10.1007/s00408-020-00361-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/08/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE The immuno-nutritional status is closely related to the prognosis in many cancers. Controlling nutritional status (CONUT) score is a new parameter that reflects the immuno-nutritional status and is prognostic in some cancers. However, the prognostic significance of the CONUT score in small cell lung cancer (SCLC) is unknown. We aimed to demonstrate the prognostic significance of the CONUT score in patients with SCLC. METHODS Two hundred sixteen patients who were followed up with SCLC were included in the study retrospectively. According to the receiver operating characteristic (ROC) curve analysis, the optimal cutoff values were determined for the CONUT score, and the patients were divided into low (< 2) and high (≥ 2) CONUT groups. Neutrophil-lymphocyte ratio (NLR), systemic immune-inflammation index (SII), and prognostic nutritional index (PNI) were grouped based on a cutoff point 2.84, 626, and 46.1, respectively. Cox regression analyses were used to assess their prognostic values for progression-free survival (PFS) and overall survival (OS). RESULTS The high CONUT group had significantly worse PFS and OS than the low CONUT group (p < 0.001, p < 0.001). In univariate analysis, stage, prophylactic cranial irradiation, extrapulmonary lesion, PNI, body mass index, CONUT score were found to be significant for both PFS and OS. In multivariate analysis, only CONUT score and stage were found as independent prognostic factors for both PFS (p: 0.018, p: 0.046) and OS (p: 0.038, p: 0.006). CONCLUSION The CONUT score at the time of diagnosis is an independent prognostic parameter that predicts recurrence and survival times in SCLC.
Collapse
Affiliation(s)
- Ali Yılmaz
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey.
| | - Salim Başol Tekin
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | - Mehmet Bilici
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | - Hatice Yılmaz
- Department Of Medical Oncology, Adnan Menderes University Faculty of Medicine, Aydın, Turkey
| |
Collapse
|
19
|
Cristea S, Coles GL, Hornburg D, Gershkovitz M, Arand J, Cao S, Sen T, Williamson SC, Kim JW, Drainas AP, He A, Cam LL, Byers LA, Snyder MP, Contrepois K, Sage J. The MEK5-ERK5 Kinase Axis Controls Lipid Metabolism in Small-Cell Lung Cancer. Cancer Res 2020; 80:1293-1303. [PMID: 31969375 PMCID: PMC7073279 DOI: 10.1158/0008-5472.can-19-1027] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 12/13/2019] [Accepted: 01/13/2020] [Indexed: 12/31/2022]
Abstract
Small-cell lung cancer (SCLC) is an aggressive form of lung cancer with dismal survival rates. While kinases often play key roles driving tumorigenesis, there are strikingly few kinases known to promote the development of SCLC. Here, we investigated the contribution of the MAPK module MEK5-ERK5 to SCLC growth. MEK5 and ERK5 were required for optimal survival and expansion of SCLC cell lines in vitro and in vivo. Transcriptomics analyses identified a role for the MEK5-ERK5 axis in the metabolism of SCLC cells, including lipid metabolism. In-depth lipidomics analyses showed that loss of MEK5/ERK5 perturbs several lipid metabolism pathways, including the mevalonate pathway that controls cholesterol synthesis. Notably, depletion of MEK5/ERK5 sensitized SCLC cells to pharmacologic inhibition of the mevalonate pathway by statins. These data identify a new MEK5-ERK5-lipid metabolism axis that promotes the growth of SCLC. SIGNIFICANCE: This study is the first to investigate MEK5 and ERK5 in SCLC, linking the activity of these two kinases to the control of cell survival and lipid metabolism.
Collapse
Affiliation(s)
- Sandra Cristea
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Garry L Coles
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Daniel Hornburg
- Department of Genetics, Stanford University, Stanford, California
| | - Maya Gershkovitz
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Julia Arand
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Siqi Cao
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Triparna Sen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stuart C Williamson
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Jun W Kim
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Alexandros P Drainas
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Andrew He
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Laurent Le Cam
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Lauren Averett Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael P Snyder
- Department of Genetics, Stanford University, Stanford, California
| | - Kévin Contrepois
- Department of Genetics, Stanford University, Stanford, California
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, California.
- Department of Genetics, Stanford University, Stanford, California
| |
Collapse
|
20
|
Alogaili F, Chinnarasu S, Jaeschke A, Kranias EG, Hui DY. Hepatic HAX-1 inactivation prevents metabolic diseases by enhancing mitochondrial activity and bile salt export. J Biol Chem 2020; 295:4631-4646. [PMID: 32079675 DOI: 10.1074/jbc.ra119.012361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/04/2020] [Indexed: 12/26/2022] Open
Abstract
Increasing hepatic mitochondrial activity through pyruvate dehydrogenase and elevating enterohepatic bile acid recirculation are promising new approaches for metabolic disease therapy, but neither approach alone can completely ameliorate disease phenotype in high-fat diet-fed mice. This study showed that diet-induced hepatosteatosis, hyperlipidemia, and insulin resistance can be completely prevented in mice with liver-specific HCLS1-associated protein X-1 (HAX-1) inactivation. Mechanistically, we showed that HAX-1 interacts with inositol 1,4,5-trisphosphate receptor-1 (InsP3R1) in the liver, and its absence reduces InsP3R1 levels, thereby improving endoplasmic reticulum-mitochondria calcium homeostasis to prevent excess calcium overload and mitochondrial dysfunction. As a result, HAX-1 ablation activates pyruvate dehydrogenase and increases mitochondria utilization of glucose and fatty acids to prevent hepatosteatosis, hyperlipidemia, and insulin resistance. In contrast to the reduction of InsP3R1 levels, hepatic HAX-1 deficiency increases bile salt exporter protein levels, thereby promoting enterohepatic bile acid recirculation, leading to activation of bile acid-responsive genes in the intestinal ileum to augment insulin sensitivity and of cholesterol transport genes in the liver to suppress hyperlipidemia. The dual mechanisms of increased mitochondrial respiration and enterohepatic bile acid recirculation due to improvement of endoplasmic reticulum-mitochondria calcium homeostasis with hepatic HAX-1 inactivation suggest that this may be a potential therapeutic target for metabolic disease intervention.
Collapse
Affiliation(s)
- Fawzi Alogaili
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Sivaprakasam Chinnarasu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| |
Collapse
|
21
|
Min BK, Oh CJ, Park S, Lee JM, Go Y, Park BY, Kang HJ, Kim DW, Kim JE, Yoo EK, Kim HE, Kim MJ, Jeon YH, Kim YH, Lee CH, Jeon JH, Lee IK. Therapeutic effect of dichloroacetate against atherosclerosis via hepatic FGF21 induction mediated by acute AMPK activation. Exp Mol Med 2019; 51:1-12. [PMID: 31570705 PMCID: PMC6802614 DOI: 10.1038/s12276-019-0315-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
Dyslipidemia-induced atherosclerosis, which has a risk of high morbidity and mortality, can be alleviated by metabolic activation associated with mitochondrial function. The effect of dichloroacetate (DCA), a general pyruvate dehydrogenase kinase (PDK) inhibitor, on in vivo energy expenditure in ApoE-/- mice fed a western diet (WD) has not yet been investigated. WD-fed ApoE-/- mice developed atherosclerotic plaques and hyperlipidemia along with obesity, which were significantly ameliorated by DCA administration. Increased oxygen consumption was associated with heat production in the DCA-treated group, with no change in food intake or physical activity compared with those of the control. These processes were correlated with the increased gene expression of Dio2 and Ucp-1, which represents brown adipose tissue (BAT) activation, in both WD-induced atherosclerosis and high-fat-induced obesity models. In addition, we found that DCA stimulated hepatic fibroblast growth factor 21 (Fgf21) mRNA expression, which might be important for lowering lipid levels and insulin sensitization via BAT activation, in a dose- and time-dependent manner associated with serum FGF21 levels. Interestingly, Fgf21 mRNA expression was mediated in an AMP-activated protein kinase (AMPK)-dependent manner within several minutes after DCA treatment independent of peroxisome proliferator-activated receptor alpha (PPARα). Taken together, the results suggest that enhanced glucose oxidation by DCA protects against atherosclerosis by inducing hepatic FGF21 expression and BAT activation, resulting in augmented energy expenditure for heat generation.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/genetics
- AMP-Activated Protein Kinases/metabolism
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Animals
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/drug therapy
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Cardiovascular Agents/pharmacology
- Dichloroacetic Acid/pharmacology
- Diet, Western/adverse effects
- Dyslipidemias/drug therapy
- Dyslipidemias/etiology
- Dyslipidemias/genetics
- Dyslipidemias/pathology
- Energy Metabolism/drug effects
- Enzyme Inhibitors/pharmacology
- Fibroblast Growth Factors/agonists
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Gene Expression Regulation
- Iodide Peroxidase/genetics
- Iodide Peroxidase/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitochondria/drug effects
- Mitochondria/metabolism
- Obesity/drug therapy
- Obesity/etiology
- Obesity/genetics
- Obesity/pathology
- Oxygen Consumption/drug effects
- PPAR alpha/genetics
- PPAR alpha/metabolism
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/etiology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/pathology
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Uncoupling Protein 1/genetics
- Uncoupling Protein 1/metabolism
- Iodothyronine Deiodinase Type II
Collapse
Affiliation(s)
- Byong-Keol Min
- Department of Biomedical Science, Graduate School and BK21 plus KNU Biomedical Convergence Programs, Daegu, South Korea
| | - Chang Joo Oh
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Sungmi Park
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Ji-Min Lee
- Department of Biomedical Science, Graduate School and BK21 plus KNU Biomedical Convergence Programs, Daegu, South Korea
| | - Younghoon Go
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| | - Bo-Yoon Park
- Department of Biomedical Science, Graduate School and BK21 plus KNU Biomedical Convergence Programs, Daegu, South Korea
| | - Hyeon-Ji Kang
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Dong Wook Kim
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Jeong-Eun Kim
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Eun Kyung Yoo
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Hui Eon Kim
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Mi-Jin Kim
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jae-Han Jeon
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - In-Kyu Lee
- Department of Biomedical Science, Graduate School and BK21 plus KNU Biomedical Convergence Programs, Daegu, South Korea.
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea.
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
22
|
Metabolic stress controls mutant p53 R248Q stability in acute myeloid leukemia cells. Sci Rep 2019; 9:5637. [PMID: 30948782 PMCID: PMC6449403 DOI: 10.1038/s41598-019-42220-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/26/2019] [Indexed: 01/03/2023] Open
Abstract
Eliminating mutant p53 (mt p53) protein could be a useful strategy to treat mt p53 tumors and potentially improve the prognosis of cancer patients. In this study, we unveil different mechanisms that eliminate p53-R248Q, one of the most frequent mutants found in human cancers. We show that the Hsp90 inhibitor 17-AAG eliminates R248Q by stimulating macroautophagy under normal growth conditions. Metabolic stress induced by the pyruvate dehydrogenase kinase-1 (PDK1) inhibitor dichloroacetate (DCA) inhibits the macroautophagy pathway. This induces the accumulation of R248Q, which in addition further inhibits macroautophagy. Combination of DCA and 17-AAG further decreases the autophagy flux compared to DCA alone. Despite this, this co-treatment strongly decreases R248Q levels. In this situation of metabolic stress, 17-AAG induces the binding of p53-R248Q to Hsc70 and the activation of Chaperone-Mediated Autophagy (CMA), leading to higher R248Q degradation than in non-stress conditions. Thus, different metabolic contexts induce diverse autophagy mechanisms that degrade p53-R248Q, and under metabolic stress, its degradation is CMA-mediated. Hence, we present different strategies to eliminate this mutant and provide new evidence of the crosstalk between macroautophagy and CMA and their potential use to target mutant p53.
Collapse
|
23
|
Serebrovska TV, Portnychenko AG, Portnichenko VI, Xi L, Egorov E, Antoniuk-Shcheglova I, Naskalova S, Shatylo VB. Effects of intermittent hypoxia training on leukocyte pyruvate dehydrogenase kinase 1 (PDK-1) mRNA expression and blood insulin level in prediabetes patients. Eur J Appl Physiol 2019; 119:813-823. [PMID: 30701312 DOI: 10.1007/s00421-019-04072-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 01/04/2019] [Indexed: 02/08/2023]
Abstract
PURPOSE Intermittent hypoxia training/treatment (IHT) is an emerging therapeutic approach to alleviate chronic diseases, such as diabetes. The present study investigated the effects of IHT on blood leucocyte pyruvate dehydrogenase kinase 1 (PDK-1) mRNA expression and its relationship with the changes in blood insulin level. METHODS Seven adult healthy volunteers and 11 prediabetic patients participated in this study. A 3-week course of IHT consisted of a 40-min session of 4 cycles of 5-min 12% O2 and 5-min room air breathing per day, 3 sessions per week for 3 weeks (i.e., total 9 sessions of IHT). Plasma insulin levels and leukocyte PDK-1 mRNA expression were determined at various time points either under fasting condition or following oral glucose tolerance test (OGTT). Correlation between the IHT-induced changes in PDK-1 mRNA and insulin or glucose levels in the same serological samples was analyzed. RESULTS At pre-IHT baseline, PDK-1 mRNA expression was two times higher in prediabetes than control subjects. IHT resulted in significant augmentation in PDK-1 mRNA expression (> twofold) in prediabetes at the end of 3-week IHT and remained elevated 1 month after IHT, which was correlated with a significantly reduced insulin release and lower blood glucose after glucose loading with OGTT. CONCLUSION IHT can trigger beneficial effects in normalizing blood insulin levels in prediabetic patients under oral glucose load, which were closely correlated with an enhanced mRNA expression of PDK-1 in leukocytes. Further clinical trials are warranted to validate the utility of IHT as a non-invasive complementary therapy against diabetes-associated pathologies.
Collapse
Affiliation(s)
| | - Alla G Portnychenko
- Bogomoletz Institute of Physiology, Kiev, 01024, Ukraine
- ICAMER, National Academy of Sciences of Ukraine, Kiev, 03680, Ukraine
| | - Vladimir I Portnichenko
- Bogomoletz Institute of Physiology, Kiev, 01024, Ukraine
- ICAMER, National Academy of Sciences of Ukraine, Kiev, 03680, Ukraine
| | - Lei Xi
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020C, Richmond, VA, 23298, USA.
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, 610041, China.
| | - Egor Egorov
- CELLGYM Technologies GmbH, 10623, Berlin, Germany
| | | | | | - Valeriy B Shatylo
- D.F. Chebotarev State Institute of Gerontology, Kiev, 04114, Ukraine
| |
Collapse
|
24
|
Mitochondrial Complex I activity signals antioxidant response through ERK5. Sci Rep 2018; 8:7420. [PMID: 29743487 PMCID: PMC5943249 DOI: 10.1038/s41598-018-23884-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/21/2018] [Indexed: 11/29/2022] Open
Abstract
Oxidative phosphorylation (OXPHOS) generates ROS as a byproduct of mitochondrial complex I activity. ROS-detoxifying enzymes are made available through the activation of their antioxidant response elements (ARE) in their gene promoters. NRF2 binds to AREs and induces this anti-oxidant response. We show that cells from multiple origins performing OXPHOS induced NRF2 expression and its transcriptional activity. The NRF2 promoter contains MEF2 binding sites and the MAPK ERK5 induced MEF2-dependent NRF2 expression. Blocking OXPHOS in a mouse model decreased Erk5 and Nrf2 expression. Furthermore, fibroblasts derived from patients with mitochondrial disorders also showed low expression of ERK5 and NRF2 mRNAs. Notably, in cells lacking functional mitochondrial complex I activity OXPHOS did not induce ERK5 expression and failed to generate this anti-oxidant response. Complex I activity induces ERK5 expression through fumarate accumulation. Eukaryotic cells have evolved a genetic program to prevent oxidative stress directly linked to OXPHOS and not requiring ROS.
Collapse
|
25
|
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: 2.4] [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
|