1
|
Costa FP, Wiedenmann B, Schöll E, Tuszynski J. Emerging cancer therapies: targeting physiological networks and cellular bioelectrical differences with non-thermal systemic electromagnetic fields in the human body - a comprehensive review. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1483401. [PMID: 39720338 PMCID: PMC11666389 DOI: 10.3389/fnetp.2024.1483401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024]
Abstract
A steadily increasing number of publications support the concept of physiological networks, and how cellular bioelectrical properties drive cell proliferation and cell synchronization. All cells, especially cancer cells, are known to possess characteristic electrical properties critical for physiological behavior, with major differences between normal and cancer cell counterparts. This opportunity can be explored as a novel treatment modality in Oncology. Cancer cells exhibit autonomous oscillations, deviating from normal rhythms. In this context, a shift from a static view of cellular processes is required for a better understanding of the dynamic connections between cellular metabolism, gene expression, cell signaling and membrane polarization as states in constant flux in realistic human models. In oncology, radiofrequency electromagnetic fields have produced sustained responses and improved quality of life in cancer patients with minimal side effects. This review aims to show how non-thermal systemic radiofrequency electromagnetic fields leads to promising therapeutic responses at cellular and tissue levels in humans, supporting this newly emerging cancer treatment modality with early favorable clinical experience specifically in advanced cancer.
Collapse
Affiliation(s)
| | | | - Eckehard Schöll
- Institut für Theoretische Physik, Technische Universität Berlin, Berlin, Germany
| | - Jack Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Turin, Italy
- Department of Data Science and Engineering, The Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
2
|
Kouba S, Demaurex N. S-acylation of Ca 2+ transport proteins in cancer. Chronic Dis Transl Med 2024; 10:263-280. [PMID: 39429488 PMCID: PMC11483607 DOI: 10.1002/cdt3.146] [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: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 10/22/2024] Open
Abstract
Alterations in cellular calcium (Ca2+) signals have been causally associated with the development and progression of human cancers. Cellular Ca2+ signals are generated by channels, pumps, and exchangers that move Ca2+ ions across membranes and are decoded by effector proteins in the cytosol or in organelles. S-acylation, the reversible addition of 16-carbon fatty acids to proteins, modulates the activity of Ca2+ transporters by altering their affinity for lipids, and enzymes mediating this reversible post-translational modification have also been linked to several types of cancers. Here, we compile studies reporting an association between Ca2+ transporters or S-acylation enzymes with specific cancers, as well as studies reporting or predicting the S-acylation of Ca2+ transporters. We then discuss the potential role of S-acylation in the oncogenic potential of a subset of Ca2+ transport proteins involved in cancer.
Collapse
Affiliation(s)
- Sana Kouba
- Department of Cell Physiology and MetabolismCentre Médical Universitaire, University of GenevaGenevaSwitzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and MetabolismCentre Médical Universitaire, University of GenevaGenevaSwitzerland
| |
Collapse
|
3
|
Stejerean-Todoran I, Gibhardt CS, Bogeski I. Calcium signals as regulators of ferroptosis in cancer. Cell Calcium 2024; 124:102966. [PMID: 39504596 DOI: 10.1016/j.ceca.2024.102966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
The field of ferroptosis research has grown exponentially since this form of cell death was first identified over a decade ago. Ferroptosis, an iron- and ROS-dependent type of cell death, is controlled by various metabolic pathways, including but not limited to redox and calcium (Ca2+) homeostasis, iron fluxes, mitochondrial function and lipid metabolism. Importantly, therapy-resistant tumors are particularly susceptible to ferroptotic cell death, rendering ferroptosis a promising therapeutic strategy against numerous malignancies. Calcium signals are important regulators of both cancer progression and cell death, with recent studies indicating their involvement in ferroptosis. Cells undergoing ferroptosis are characterized by plasma membrane rupture and the formation of nanopores, which facilitate influx of ions such as Ca2+ into the affected cells. Furthermore, mitochondrial Ca²⁺ levels have been implicated in directly influencing the cellular response to ferroptosis. Despite the remarkable progress made in the field, our understanding of the contribution of Ca2+ signals to ferroptosis remains limited. Here, we summarize key connections between Ca²⁺ signaling and ferroptosis in cancer pathobiology and discuss their potential therapeutic significance.
Collapse
Affiliation(s)
- Ioana Stejerean-Todoran
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
4
|
Bigham NP, Novorolsky RJ, Davis KR, Zou H, MacMillan SN, Stevenson MJ, Robertson GS, Wilson JJ. Supramolecular delivery of dinuclear ruthenium and osmium MCU inhibitors. Inorg Chem Front 2024; 11:5064-5079. [PMID: 39113903 PMCID: PMC11301636 DOI: 10.1039/d4qi01102c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024]
Abstract
The transmembrane protein known as the mitochondrial calcium uniporter (MCU) mediates the influx of calcium ions (Ca2+) into the mitochondrial matrix. An overload of mitochondrial Ca2+ ( m Ca2+) is directly linked to damaging effects in pathological conditions. Therefore, inhibitors of the MCU are important chemical biology tools and therapeutic agents. Here, two new analogues of previously reported Ru- and Os-based MCU inhibitors Ru265 and Os245, of the general formula [(C10H15CO2)M(NH3)4(μ-N)M(NH3)4(O2CC10H15)](CF3SO3)3, where M = Ru (1) or Os (2), are reported. These analogues bear adamantane functional groups, which were installed to act as guests for the host molecule cucurbit-[7]-uril (CB[7]). These complexes were characterized and analyzed for their efficiency as guests for CB[7]. As shown through a variety of spectroscopic techniques, each adamantane ligand is encapsulated into one CB[7], affording a supramolecular complex of 1 : 2 stoichiometry. The biological effects of these compounds in the presence and absence of two equiv. CB[7] were assessed. Both complexes 1 and 2 exhibit enhanced cellular uptake compared to the parent compounds Ru265 and Os245, and their uptake is increased further in the presence of CB[7]. Compared to Ru265 and Os245, 1 and 2 are less potent as m Ca2+ uptake inhibitors in permeabilized cell models. However, in intact cell systems, 1 and 2 inhibit the MCU at concentrations as low as 1 μM, marking an advantage over Ru265 and Os245 which require an order of magnitude higher doses for similar biological effects. The presence of CB[7] did not affect the inhibitory properties of 1 and 2. Experiments in primary cortical neurons showed that 1 and 2 can elicit protective effects against oxygen-glucose deprivation at lower doses than those required for Ru265 or Os245. At low concentrations, the protective effects of 1 were modulated by CB[7], suggesting that supramolecular complex formation can play a role in these biological conditions. The in vivo biocompatibility of 1 was investigated in mice. The intraperitoneal administration of these compounds and their CB[7] complexes led to time-dependent induction of seizures with no protective effects elicited by CB[7]. This work demonstrates the potential for supramolecular interactions in the development of MCU inhibitors.
Collapse
Affiliation(s)
- Nicholas P Bigham
- Department of Chemistry and Chemical Biology, Cornell University Ithaca NY 14853 USA
| | - Robyn J Novorolsky
- Department of Pharmacology, Faculty of Medicine, Dalhousie University 6th Floor Sir Charles Tupper Medical Building Halifax B3H 4R2 Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Sciences Research Institute Halifax NS B3H 4R2 Canada
| | - Keana R Davis
- Department of Chemistry, University of San Francisco San Francisco CA 94117 USA
| | - Haipei Zou
- Department of Chemistry and Chemical Biology, Cornell University Ithaca NY 14853 USA
- Department of Chemistry & Biochemistry, University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University Ithaca NY 14853 USA
| | - Michael J Stevenson
- Department of Chemistry, University of San Francisco San Francisco CA 94117 USA
| | - George S Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University 6th Floor Sir Charles Tupper Medical Building Halifax B3H 4R2 Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Sciences Research Institute Halifax NS B3H 4R2 Canada
- Department of Psychiatry, Faculty of Medicine, Dalhousie University Halifax NS B3H 2E2 Canada
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University Ithaca NY 14853 USA
- Department of Chemistry & Biochemistry, University of California Santa Barbara Santa Barbara CA 93106 USA
| |
Collapse
|
5
|
Huang SQ, Cao KX, Wang CL, Chen PL, Chen YX, Zhang YT, Yu SH, Bai ZX, Guo S, Liao MX, Li QW, Zhang GQ, He J, Xu YM. Decreasing mitochondrial fission ameliorates HIF-1α-dependent pathological retinal angiogenesis. Acta Pharmacol Sin 2024; 45:1438-1450. [PMID: 38565961 PMCID: PMC11192750 DOI: 10.1038/s41401-024-01262-3] [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: 08/27/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Angiogenesis plays a critical role in many pathological processes, including irreversible blindness in eye diseases such as retinopathy of prematurity. Endothelial mitochondria are dynamic organelles that undergo constant fusion and fission and are critical signalling hubs that modulate angiogenesis by coordinating reactive oxygen species (ROS) production and calcium signalling and metabolism. In this study, we investigated the role of mitochondrial dynamics in pathological retinal angiogenesis. We showed that treatment with vascular endothelial growth factor (VEGF; 20 ng/ml) induced mitochondrial fission in HUVECs by promoting the phosphorylation of dynamin-related protein 1 (DRP1). DRP1 knockdown or pretreatment with the DRP1 inhibitor Mdivi-1 (5 μM) blocked VEGF-induced cell migration, proliferation, and tube formation in HUVECs. We demonstrated that VEGF treatment increased mitochondrial ROS production in HUVECs, which was necessary for HIF-1α-dependent glycolysis, as well as proliferation, migration, and tube formation, and the inhibition of mitochondrial fission prevented VEGF-induced mitochondrial ROS production. In an oxygen-induced retinopathy (OIR) mouse model, we found that active DRP1 was highly expressed in endothelial cells in neovascular tufts. The administration of Mdivi-1 (10 mg·kg-1·d-1, i.p.) for three days from postnatal day (P) 13 until P15 significantly alleviated pathological angiogenesis in the retina. Our results suggest that targeting mitochondrial fission may be a therapeutic strategy for proliferative retinopathies and other diseases that are dependent on pathological angiogenesis.
Collapse
Affiliation(s)
- Shu-Qi Huang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kai-Xiang Cao
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Cai-Ling Wang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Pei-Ling Chen
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi-Xin Chen
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Ting Zhang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shi-Hui Yu
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zai-Xia Bai
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuai Guo
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mu-Xi Liao
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, China
| | - Qiao-Wen Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511520, China
| | - Guo-Qi Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511520, China.
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, China.
| | - Yi-Ming Xu
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
6
|
Tian P, Du D, Yang L, Zhou N, Tao L. Lentinan mitigates pemetrexed chemoresistance by the PI3K/Akt pathway in non-small cell lung cancer. Cell Biochem Biophys 2024; 82:1421-1431. [PMID: 38750384 DOI: 10.1007/s12013-024-01296-8] [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] [Accepted: 04/24/2024] [Indexed: 08/25/2024]
Abstract
Pemetrexed is a folate analog metabolic inhibitor that is given for therapy of non-small cell lung cancer (NSCLC). Drug resistance affects the efficacy of pemetrexed in NSCLC. Lentinan is a polysaccharide extracted from Shiitake mushrooms which has antitumor roles in multiple cancers, including lung cancer. However, the effects of lentinan on pemetrexed resistance in NSCLC remain unclear. In present study, The pemetrexed-resistant NSCLC cells were established and exposed to pemetrexed and lentinan. Oxidative stress was investigated via mitochondrial membrane potential (JC-1 staining), levels of MDA and SOD.The phosphorylation and total of PI3K and Akt levels were actuated using specific activator 740Y-P and measured through western blot. We observed that Lentinan decreased IC50 of pemetrexed in resistant NSCLC cells. Lentinan aggravated pemetrexed-induced proliferation inhibition of resistant NSCLC cells via reducing PCNA levels. Lentinan exacerbated pemetrexed-triggered oxidative stress through increasing ROS and MDA levels, and reducing mitochondrial membrane potential and SOD levels. Lentinan inhibited PI3K/Akt signaling activation in pemetrexed-treated cells. Activated PI3K/Akt pathway using activator 740Y-P reversed the effects of lentinan on pemetrexed-mediated proliferation inhibition and oxidative stress. Our findings uncover that Lentinan mitigates pemetrexed resistance in NSCLC through inhibiting cell proliferation and inducing oxidative stress by suppressing PI3K/Akt signaling.
Collapse
Affiliation(s)
- Ping Tian
- School of Medical, Xinyang Vocational and Technical College, Xinyang, 464000, Henan, China
| | - Dajun Du
- Department of Cancer Surgery, Xinyang Central Hospital, Xinyang, 464000, Henan, China
| | - Li Yang
- School of Inspection, Xinyang Vocational and Technical College, Xinyang, 464000, Henan, China
| | - Nan Zhou
- Department of Medical Oncology, Xinyang Central Hospital, Xinyang, 464000, Henan, China
| | - Ling Tao
- School of Inspection, Xinyang Vocational and Technical College, Xinyang, 464000, Henan, China.
| |
Collapse
|
7
|
Zhang L, Ren C, Liu J, Huang S, Wu C, Zhang J. Development and therapeutic implications of small molecular inhibitors that target calcium-related channels in tumor treatment. Drug Discov Today 2024; 29:103995. [PMID: 38670255 DOI: 10.1016/j.drudis.2024.103995] [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: 02/03/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Calcium ion dysregulation exerts profound effects on various physiological activities such as tumor proliferation, migration, and drug resistance. Calcium-related channels play a regulatory role in maintaining calcium ion homeostasis, with most channels being highly expressed in tumor cells. Additionally, these channels serve as potential drug targets for the development of antitumor medications. In this review, we first discuss the current research status of these pathways, examining how they modulate various tumor functions such as epithelial-mesenchymal transition (EMT), metabolism, and drug resistance. Simultaneously, we summarize the recent progress in the study of novel small-molecule drugs over the past 5 years and their current status.
Collapse
Affiliation(s)
- Linxi Zhang
- China Medical University-Queen's University of Belfast Joint College, China Medical University, Shenyang 110000, Liaoning, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu 611130, China
| | - Jiao Liu
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu 611130, China
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Chengyong Wu
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
8
|
Calbiague-Garcia V, Chen Y, Cádiz B, Tapia F, Paquet-Durand F, Schmachtenberg O. Extracellular lactate as an alternative energy source for retinal bipolar cells. J Biol Chem 2024; 300:106794. [PMID: 38403245 PMCID: PMC10966802 DOI: 10.1016/j.jbc.2024.106794] [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: 12/22/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
Retinal bipolar and amacrine cells receive visual information from photoreceptors and participate in the first steps of image processing in the retina. Several studies have suggested the operation of aerobic glycolysis and a lactate shuttle system in the retina due to the high production of this metabolite under aerobic conditions. However, whether bipolar cells form part of this metabolic circuit remains unclear. Here, we show that the monocarboxylate transporter 2 is expressed and functional in inner retinal neurons. Additionally, we used genetically encoded FRET nanosensors to demonstrate the ability of inner retinal neurons to consume extracellular lactate as an alternative to glucose. In rod bipolar cells, lactate consumption allowed cells to maintain the homeostasis of ions and electrical responses. We also found that lactate synthesis and transporter inhibition caused functional alterations and an increased rate of cell death. Overall, our data shed light on a notable but still poorly understood aspect of retinal metabolism.
Collapse
Affiliation(s)
- Victor Calbiague-Garcia
- PhD Program in Neuroscience, Universidad de Valparaíso, Valparaíso, Chile; CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile.
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Bárbara Cádiz
- CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Tapia
- CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile
| | | | | |
Collapse
|
9
|
Xu A, Wang Y, Luo D, Xia Y, Xue H, Yao H, Li S. By regulating the IP3R/GRP75/VDAC1 complex to restore mitochondrial dynamic balance, selenomethionine reduces lipopolysaccharide-induced neuronal apoptosis. J Cell Physiol 2024; 239:e31190. [PMID: 38219075 DOI: 10.1002/jcp.31190] [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: 10/23/2023] [Revised: 12/09/2023] [Accepted: 12/23/2023] [Indexed: 01/15/2024]
Abstract
Selenium (Se), as one of the essential trace elements, plays an anti-inflammatory, antioxidation, and immune-enhancing effect in the body. In addition, Se can also improve nervous system damage induced by various factors. Earlier studies have described the important role of mitochondrial dynamic imbalance in lipopolysaccharide (LPS)-induced nerve injury. The inositol 1,4,5-triphosphate receptor (IP3R)/glucose-regulated protein 75 (GRP75)/voltage-dependent anion channel 1 (VDAC1) complex is considered to be the key to regulating mitochondrial dynamics. However, it is not clear whether Selenomethionine (SeMet) has any influence on the IP3R/GRP75/VDAC1 complex. Therefore, the aim of this investigation was to determine whether SeMet can alleviate LPS-induced brain damage and to elucidate the function of the IP3R/GRP75/VDAC1 complex in it. We established SeMet and/or LPS exposure models in vivo and in vitro using laying hens and primary chicken nerve cells. We noticed that SeMet reversed endoplasmic reticulum stress (ERS) and the imbalance in mitochondrial dynamics and significantly prevented the occurrence of neuronal apoptosis. We made this finding by morphological observation of the brain tissue of laying hens and the detection of related genes such as ERS, the IP3R/GRP75/VDAC1 complex, calcium signal (Ca2+), mitochondrial dynamics, and apoptosis. Other than that, we also discovered that the IP3R/GRP75/VDAC1 complex was crucial in controlling Ca2+ transport between the endoplasmic reticulum and the mitochondrion when SeMet functions as a neuroprotective agent. In summary, our results revealed the specific mechanism by which SeMet alleviated LPS-induced neuronal apoptosis for the first time. As a consequence, SeMet has great potential in the treatment and prevention of neurological illnesses (like neurodegenerative diseases).
Collapse
Affiliation(s)
- Anqi Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yixuan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Dongliu Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yu Xia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hua Xue
- National Selenium-Rich Product Quality Supervision and Inspection Center, Enshi, People's Republic of China
| | - Haidong Yao
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
10
|
Wang K, Du Y, Li P, Guan C, Zhou M, Wu L, Liu Z, Huang Z. Nanoplastics causes heart aging/myocardial cell senescence through the Ca 2+/mtDNA/cGAS-STING signaling cascade. J Nanobiotechnology 2024; 22:96. [PMID: 38448951 PMCID: PMC10918962 DOI: 10.1186/s12951-024-02375-x] [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: 09/05/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Nanoplastics (NPs) are now a new class of pollutants widely present in the soil, atmosphere, freshwater and marine environments. Nanoplastics can rapidly penetrate cell membranes and accumulate in human tissues and organs, thus posing a potential threat to human health. The heart is the main power source of the body. But up to now, the toxicological effects of long-term exposure to nanoplastics on the heart has not been revealed yet. RESULTS We evaluated the effects of long term exposure of nanoplastics on cardiac cell/tissue in vitro and in vivo model. Furthermore, we explored the molecular mechanism by which nanoplastics exposure causes myocardial cell senescence. Immunohistochemistry, indirect immunofluorescence and ELISA were performed to detect the effects of nanoplastics on heart aging. We found that nanoplastics were able to induce significant cardiac aging through a series of biochemical assays in vivo. In vitro, the effects of nanoplastics on cardiac cell were investigated, and found that nanoplastics were able to internalize into cardiomyocytes in time and dose-dependant manner. Further biochemical analysis showed that nanoplastics induces cardiomyocytes senescence by detecting a series of senescence marker molecules. Molecular mechanism research shows that nanoplastics may cause mitochondrial destabilization by inducing oxidative stress, which leads to the leakage of mtDNA from mitochondria into the cytoplasm, and then cytoplasm-localized mt-DNA activates the cGAS-STING signaling pathway and promotes inflammation response, ultimately inducing cardiomyocytes senescence. CONCLUSIONS In this work, we found that nanoplastics exposure induces premature aging of heart. Current research also reveals the molecular mechanism by which nanoplastics induces cardiomyocyte senescence. This study laid the foundation for further studying the potential harm of nanoplastics exposure on heart.
Collapse
Affiliation(s)
- Kaihao Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yipeng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peixin Li
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chang Guan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lanlan Wu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zengfu Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zheng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Heiliczer S, Yanko R, Sharav Y, Aframian DJ, Klutstein M, Wilensky A, Haviv Y. Oxidative stress-mediated proapoptosis signaling: A novel theory on the mechanism underlying the pathogenesis of burning mouth syndrome. J Am Dent Assoc 2024; 155:258-267. [PMID: 37966403 DOI: 10.1016/j.adaj.2023.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Burning mouth syndrome (BMS) is a chronic oral pain disorder characterized by a generalized burning sensation in the oral mucosa without apparent medical or dental causes. Despite various hypotheses proposed to explain BMS pathogenesis, a clear understanding of the cellular-level events and associated histologic and molecular findings is lacking. Advancing our understanding of BMS pathogenesis could facilitate the development of more targeted therapeutic interventions. TYPES OF STUDIES REVIEWED The authors conducted an extensive literature search and review of cellular mechanisms, focusing on evidence-based data that support a comprehensive hypothesis for BMS pathogenesis. The authors explored novel and detailed mechanisms that may account for the characteristic features of BMS. RESULTS The authors proposed that BMS symptoms arise from the uncontrolled activation of proapoptotic transmembrane calcium permeable channels expressed in intraoral mucosal nerve fibers. Elevated levels of reactive oxygen species or dysfunctional antiapoptosis pathways may lead to uncontrolled oxidative stress-mediated apoptosis signaling, resulting in upregulation of transmembrane transient receptor potential vanilloid type 1 and P2X 3 calcium channels in nociceptive fibers. Activation of these channels can cause nerve terminal depolarization, leading to generation of action potentials that are centrally interpreted as pain. CONCLUSIONS AND PRACTICAL IMPLICATIONS The authors present a novel hypothesis for BMS pathogenesis, highlighting the role of proapoptotic transmembrane calcium permeable channels and oxidative stress-mediated apoptosis signaling in the development of BMS symptoms. Understanding these underlying mechanisms could provide new insights into the development of targeted therapeutic interventions for BMS. Additional research is warranted to validate this hypothesis and explore potential avenues for effective management of BMS.
Collapse
|
12
|
Cheng J, Wang X, Luo C, Mao X, Qin J, Chi Y, He B, Hao Y, Niu X, Huang B, Liu L. Effects of intracellular Ca 2+ on developmental potential and ultrastructure of cryopreserved-warmed oocyte in mouse. Cryobiology 2024; 114:104834. [PMID: 38065230 DOI: 10.1016/j.cryobiol.2023.104834] [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: 07/12/2023] [Revised: 10/15/2023] [Accepted: 12/01/2023] [Indexed: 03/22/2024]
Abstract
Maintaining appropriate intracellular calcium of oocytes is necessary to prevent ultrastructure and organelle damage caused by freezing and cryoprotectants. The present study aimed to investigate whether cryoprotectant-induced changes in the calcium concentrations of oocytes can be regulated to reduce damage to developmental potential and ultrastructure. A total of 33 mice and 1381 oocytes were used to explore the effects of intracellular calcium on the development and ultrastructures of oocytes subjected to 2-aminoethoxydiphenyl borate (2-APB) inhibition or thapsigargin (TG) stimulation. Results suggested that high levels intracellular calcium interfered with TG compromised oocyte survival (84.4 % vs. 93.4 %, p < 0.01) and blastocyst formation in fresh and cryopreservation oocytes (78.1 % vs. 86.4 %, and 60.5 % vs. 72.5 %, p < 0.05) compared with that of 2-APB pretreated oocytes in which Ca2+ was stabilized even though no differences in fertilization and cleavage was detected (p > 0.05). Examination by transmission electron microscopy indicated that the microvilli decreased and shortened, cortical granules considerably decreased in the cortex area, mitochondrial vesicles and vacuoles increased, and the proportion of vacuole mitochondria increased after oocytes were exposed to cryoprotectants. The cryopreservation-warming process deteriorated the negative effects on organelles of survival oocytes. By contrast, a low level of intracellular calcium mediated with 2-APB was supposed to contribute to the protection of organelles. These findings suggested oocyte injuries induced by cryoprotectants and low temperatures can be alleviated. More studies are necessary to confirm the relationship among Ca2+ concentration of the cytoplasm, ultrastructural injuries, and disrupted developmental potential in oocytes subjected to cryopreservation and warming.
Collapse
Affiliation(s)
- Junping Cheng
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China; College of Animal Science and Technology of Guangxi University, Nanning, 530005, China.
| | - Xiaoli Wang
- College of Animal Science and Technology of Guangxi University, Nanning, 530005, China
| | - Chan Luo
- College of Animal Science and Technology of Guangxi University, Nanning, 530005, China
| | - Xianbao Mao
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China
| | - Jie Qin
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China
| | - Yan Chi
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China
| | - Bing He
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China
| | - Yanrong Hao
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China
| | - Xiangli Niu
- Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Ben Huang
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China; College of Animal Science and Technology of Guangxi University, Nanning, 530005, China
| | - Liling Liu
- Reproductive Medical and Genetic Center, Academy of Medical Sciences of Guangxi Autonomous Region, People's Hospital of Guangxi Autonomous Region, Nanning, 530021, China.
| |
Collapse
|
13
|
Ibrahim IH. Metalloproteins and metalloproteomics in health and disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:123-176. [PMID: 38960472 DOI: 10.1016/bs.apcsb.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Metalloproteins represents more than one third of human proteome, with huge variation in physiological functions and pathological implications, depending on the metal/metals involved and tissue context. Their functions range from catalysis, bioenergetics, redox, to DNA repair, cell proliferation, signaling, transport of vital elements, and immunity. The human metalloproteomic studies revealed that many families of metalloproteins along with individual metalloproteins are dysregulated under several clinical conditions. Also, several sorts of interaction between redox- active or redox- inert metalloproteins are observed in health and disease. Metalloproteins profiling shows distinct alterations in neurodegenerative diseases, cancer, inflammation, infection, diabetes mellitus, among other diseases. This makes metalloproteins -either individually or as families- a promising target for several therapeutic approaches. Inhibitors and activators of metalloenzymes, metal chelators, along with artificial metalloproteins could be versatile in diagnosis and treatment of several diseases, in addition to other biomedical and industrial applications.
Collapse
Affiliation(s)
- Iman Hassan Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
14
|
Feng Y, Wang J, Cao J, Cao F, Chen X. Manipulating calcium homeostasis with nanoplatforms for enhanced cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230019. [PMID: 38854493 PMCID: PMC10867402 DOI: 10.1002/exp.20230019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/28/2023] [Indexed: 06/11/2024]
Abstract
Calcium ions (Ca2+) are indispensable and versatile metal ions that play a pivotal role in regulating cell metabolism, encompassing cell survival, proliferation, migration, and gene expression. Aberrant Ca2+ levels are frequently linked to cell dysfunction and a variety of pathological conditions. Therefore, it is essential to maintain Ca2+ homeostasis to coordinate body function. Disrupting the balance of Ca2+ levels has emerged as a potential therapeutic strategy for various diseases, and there has been extensive research on integrating this approach into nanoplatforms. In this review, the current nanoplatforms that regulate Ca2+ homeostasis for cancer therapy are first discussed, including both direct and indirect approaches to manage Ca2+ overload or inhibit Ca2+ signalling. Then, the applications of these nanoplatforms in targeting different cells to regulate their Ca2+ homeostasis for achieving therapeutic effects in cancer treatment are systematically introduced, including tumour cells and immune cells. Finally, perspectives on the further development of nanoplatforms for regulating Ca2+ homeostasis, identifying scientific limitations and future directions for exploitation are offered.
Collapse
Affiliation(s)
- Yanlin Feng
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Jianlin Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Agency for Science, Technology, and Research (A*STAR)Institute of Molecular and Cell BiologySingaporeSingapore
| |
Collapse
|
15
|
Duranti E, Cordani N, Villa C. Edaravone: A Novel Possible Drug for Cancer Treatment? Int J Mol Sci 2024; 25:1633. [PMID: 38338912 PMCID: PMC10855093 DOI: 10.3390/ijms25031633] [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: 12/22/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in understanding the causes and progression of tumors, cancer remains one of the leading causes of death worldwide. In light of advances in cancer therapy, there has been a growing interest in drug repurposing, which involves exploring new uses for medications that are already approved for clinical use. One such medication is edaravone, which is currently used to manage patients with cerebral infarction and amyotrophic lateral sclerosis. Due to its antioxidant and anti-inflammatory properties, edaravone has also been investigated for its potential activities in treating cancer, notably as an anti-proliferative and cytoprotective drug against side effects induced by traditional cancer therapies. This comprehensive review aims to provide updates on the various applications of edaravone in cancer therapy. It explores its potential as a standalone antitumor drug, either used alone or in combination with other medications, as well as its role as an adjuvant to mitigate the side effects of conventional anticancer treatments.
Collapse
Affiliation(s)
| | | | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.D.); (N.C.)
| |
Collapse
|
16
|
Xiao H, Ma L, Ding J, Wang H, Bi X, Tan F, Piao W. Mitochondrial Calcium Uniporter (MCU) that Modulates Mitochondrial Calcium Uptake and Facilitates Endometrial Cancer Progression through Interaction with VDAC1. Curr Cancer Drug Targets 2024; 24:354-367. [PMID: 37702230 DOI: 10.2174/1568009624666230912095526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Although endometrial cancer represents a frequently diagnosed malignancy of the female reproductive tract, we know very little about the factors that control endometrial cancer. OBJECTIVE Our study was presented to investigate the function of MCU in endometrial tumorigenesis and the molecular mechanisms involved. MATERIALS AND METHODS A total of 94 endometrial cancer patients were recruited into our cohort. MCU and VDAC1 expression was examined in tumor and normal tissues via immunohistochemistry and immunofluorescence. Associations of MCU and VDAC1 expression with clinicopathological characteristics were evaluated. After transfection with shRNA targeting MCU or full-length MCU plasmids, clone formation, wound healing, transwell and MitoTracker Red staining were separately presented in Ishikawa and RL95-2 cells. Moreover, Western blotting or immunofluorescence was utilized to examine the expression of MCU, VDAC1, Na+/Ca2+/Li+ exchanger (NCLX), and β-catenin under VDAC1 knockdown and/or MCU overexpression or knockdown. RESULTS MCU and VDAC1 expression were prominently up-regulated in endometrial cancer tissues and were significantly associated with histological grade, depth of myometrial invasion and lymph node status. MCU up-regulation enhanced clone formation, migration, and mitochondrial activity of endometrial cancer cells. The opposite results were investigated when MCU was silenced. MCU or VDAC1 silencing reduced the expression of MCU, VDAC1, NCLX, and β-catenin. Moreover, VDAC1 knockdown alleviated the promoting effect of MCU overexpression on the above proteins. CONCLUSION This investigation demonstrated that MCU-induced mitochondrial calcium uptake plays a critical role in endometrial tumorigenesis through interaction with VDAC1.
Collapse
Affiliation(s)
- Hongyan Xiao
- Department of Pathology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia Hui Autonomous Region, China
| | - Lijun Ma
- School of Electrical and Information Engineering, Department of Medical Imaging, North Minzu University, Yinchuan, 750021, Ningxia Hui Autonomous Region, China
| | - Jie Ding
- Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia Hui Autonomous Region, China
| | - Honghong Wang
- Department of Pathology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia Hui Autonomous Region, China
| | - Xiaofang Bi
- Department of Pathology, The First People's Hospital of Yinchuan, Yinchuan, 750001, Ningxia Hui Autonomous Region, China
| | - Fengmei Tan
- Department of Pathology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia Hui Autonomous Region, China
| | - Wenhua Piao
- Clinical Medical Laboratory Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, Ningxia Hui Autonomous Region, China
| |
Collapse
|
17
|
Lin CP, Wu SH, Lin TY, Chu CH, Lo LW, Kuo CC, Chang JY, Hsu SC, Ko BS, Yao M, Hsiao JK, Wang SW, Huang DM. Lysosomal-targeted doxorubicin delivery using RBC-derived vesicles to overcome drug-resistant cancer through mitochondrial-dependent cell death. Pharmacol Res 2023; 197:106945. [PMID: 37797662 DOI: 10.1016/j.phrs.2023.106945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
Multidrug resistance (MDR) is a major challenge in cancer chemotherapy. Nanoparticles as drug delivery systems (DDSs) show promise for MDR cancer therapy. However, current DDSs require sophisticated design and construction based on xenogeneic nanomaterials, evoking feasibility and biocompatibility concerns. Herein, a simple but versatile biological DDS (bDDS) composed of human red blood cell (RBC)-derived vesicles (RDVs) with excellent biocompatibility was surface-linked with doxorubicin (Dox) using glutaraldehyde (glu) to form Dox-gluRDVs that remarkably suppressed MDR in uterine sarcoma through a lysosomal-mitochondrial axis-dependent cell death mechanism. Dox-gluRDVs can efficiently deliver and accumulate Dox in lysosomes, bypassing drug efflux transporters and facilitating cellular uptake and retention of Dox in drug-resistant MES-SA/Dx5 cells. The transfer of lysosomal calcium to the mitochondria during mitochondria-lysosome contact due to lysosomal Dox accumulation may result in mitochondrial ROS overproduction, mitochondrial membrane potential loss, and activation of apoptotic signaling for the superior anti-MDR activity of Dox-gluRDVs in vitro and in vivo. This work highlights the great promise of RDVs to serve as a bDDS of Dox to overcome MDR cancers but also opens up a reliable strategy for lysosomal-mitochondrial axis-dependent cell death for fighting against other inoperable cancers.
Collapse
Affiliation(s)
- Chih-Peng Lin
- Department of Anesthesiology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan
| | - Shu-Hui Wu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Tzu-Yin Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Chia-Hui Chu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Leu-Wei Lo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Taipei Cancer Center, Taipei Medical University Hospital, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Szu-Chun Hsu
- Department of Laboratory Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan
| | - Bor-Sheng Ko
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan; Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei 106037, Taiwan
| | - Ming Yao
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City 23142, Taiwan; School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Shih-Wei Wang
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252005, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City 252005, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung 807378, Taiwan
| | - Dong-Ming Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan.
| |
Collapse
|
18
|
Liang T, Feng Z, Zhang X, Li T, Yang T, Yu L. Research progress of calcium carbonate nanomaterials in cancer therapy: challenge and opportunity. Front Bioeng Biotechnol 2023; 11:1266888. [PMID: 37811375 PMCID: PMC10551635 DOI: 10.3389/fbioe.2023.1266888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Cancer has keeping the main threat to the health of human being. Its overall survival rate has shown rare substantial progress in spite of the improving diagnostic and treatment techniques for cancer in recent years. Indeed, such classic strategies for malignant tumor as surgery, radiation and chemotherapy have been developed and bring more hope to the patients, but still been accompanied by certain limitations, which include the challenge of managing large wound sizes, systemic toxic side effects, and harmful to the healthy tissues caused by imprecise alignment with tumors in radiotherapy. Furthermore, immunotherapy exhibits a limited therapeutic effect in advanced tumors which is reported only up to 25%-30%. The combination of nanomaterials and cancer treatment offers new hope for cancer patients, demonstrating strong potential in the field of medical research. Among the extensively utilized nanomaterials, calcium carbonate nanomaterials (CCNM) exhibit a broad spectrum of biomedical applications due to their abundant availability, cost-effectiveness, and exceptional safety profile. CCNM have the potential to elevate intracellular Ca2+ levels in tumor cells, trigger the mitochondrial damage and ultimately lead to tumor cell death. Moreover, compared with other types of nanomaterials, CCNM exhibit remarkable advantages as delivery systems owing to their high loading capacity, biocompatibility and biodegradability. The purpose of this review is to provide an overview of CCNM synthesis, focusing on summarizing its diverse roles in cancer treatment and the benefits and challenges associated with CCNM in cancer therapy. Hoping to present the significance of CCNM as for the clinical application, and summarize information for the design of CCNM and other types of nanomaterials in the future.
Collapse
Affiliation(s)
- Tiantian Liang
- Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Clinical Medical Research Center, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Zongqi Feng
- Clinical Medical Research Center, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Xiao Zhang
- Clinical Medical Research Center, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Tianfang Li
- Clinical Medical Research Center, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Tingyu Yang
- Clinical Medical Research Center, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Lan Yu
- Clinical Medical Research Center, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
19
|
Ren Y, Wang R, Weng S, Xu H, Zhang Y, Chen S, Liu S, Ba Y, Zhou Z, Luo P, Cheng Q, Dang Q, Liu Z, Han X. Multifaceted role of redox pattern in the tumor immune microenvironment regarding autophagy and apoptosis. Mol Cancer 2023; 22:130. [PMID: 37563639 PMCID: PMC10413697 DOI: 10.1186/s12943-023-01831-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
The reversible oxidation-reduction homeostasis mechanism functions as a specific signal transduction system, eliciting related physiological responses. Disruptions to redox homeostasis can have negative consequences, including the potential for cancer development and progression, which are closely linked to a series of redox processes, such as adjustment of reactive oxygen species (ROS) levels and species, changes in antioxidant capacity, and differential effects of ROS on downstream cell fate and immune capacity. The tumor microenvironment (TME) exhibits a complex interplay between immunity and regulatory cell death, especially autophagy and apoptosis, which is crucially regulated by ROS. The present study aims to investigate the mechanism by which multi-source ROS affects apoptosis, autophagy, and the anti-tumor immune response in the TME and the mutual crosstalk between these three processes. Given the intricate role of ROS in controlling cell fate and immunity, we will further examine the relationship between traditional cancer therapy and ROS. It is worth noting that we will discuss some potential ROS-related treatment options for further future studies.
Collapse
Affiliation(s)
- Yuqing Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ruizhi Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Pediatric Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
20
|
Tsugeno Y, Sato T, Watanabe M, Furuhashi M, Ohguro H. Prostanoid FP and EP2 Receptor Agonists Induce Epithelial and Subepithelial Fibrogenetic Changes in Human Conjunctival Fibroblasts in Different Manners. J Ocul Pharmacol Ther 2023; 39:404-414. [PMID: 37459581 DOI: 10.1089/jop.2023.0011] [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] [Indexed: 07/20/2023] Open
Abstract
Purpose: To examine the effects of prostanoid FP and EP2 receptor agonists, PGF2α and Omidenepag (OMD), respectively, on the transforming growth factor beta (TGF-β2) induced conjunctival fibrogenesis. Methods: Two-dimension (2D) and three-dimension (3D) cultures of these fibroblasts were subjected to following analyses: (1) planar proliferation evaluated by transendothelial electron resistance (TEER) measurements, (2) real-time metabolic analyses, (3) subepithelial proliferation evaluated by 3D spheroid' size and stiffness measurements, and (4) the mRNA expression of extracellular matrix (ECM) molecules and their modulators. Results: TGF-β2 induced increase in the planar proliferation was significantly decreased or enhanced by PGF2α or OMD, respectively. The proportion of oxygen consumption required to drive ATP synthesis compared with that driving proton leakage was increased by PGF2α and OMD independently with TGF-β2. In contrast, maximal mitochondrial respiration was decreased by PGF2α and OMD, and the OMD-induced effect was further enhanced by the presence of TGF-β2. In addition, the TGF-β2 dependent increase in the glycolytic capacity was cancelled by PGF2α and/or OMD. Alternatively, subepithelial proliferation, as evidenced by the stiffness of the 3D spheroids, was substantially increased by both PGF2α and OMD, and these were differently modulated by TGF-β2. The expression of several related factors as above fluctuated among the conditions for both 2D and 3D and TGF-β2 untreated or treated cultures. Conclusion: The present findings indicate that the prostanoid FP or the EP2 receptor agonist may solely and differently induce the planar and subepithelial proliferation of HconF cells and these were also modulated by TGF-β2.
Collapse
Affiliation(s)
- Yuri Tsugeno
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Ohguro
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
21
|
Castro DTH, Leite DF, da Silva Baldivia D, Dos Santos HF, Balogun SO, da Silva DB, Carollo CA, de Picoli Souza K, Dos Santos EL. Structural Characterization and Anticancer Activity of a New Anthraquinone from Senna velutina (Fabaceae). Pharmaceuticals (Basel) 2023; 16:951. [PMID: 37513863 PMCID: PMC10385181 DOI: 10.3390/ph16070951] [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: 05/22/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
In this study, a novel compound was isolated, identified, and its chemical structure was determined from the extract of the roots of Senna velutina. In addition, we sought to evaluate the anticancer potential of this molecule against melanoma and leukemic cell lines and identify the pathways of cell death involved. To this end, a novel anthraquinone was isolated from the barks of the roots of S. velutina, analyzed by HPLC-DAD, and its molecular structure was determined by nuclear magnetic resonance (NMR). Subsequently, their cytotoxic activity was evaluated by the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) method against non-cancerous, melanoma, and leukemic cells. The migration of melanoma cells was evaluated by the scratch assay. The apoptosis process, caspase-3 activation, analysis of mitochondrial membrane potential, and measurement of ROS were evaluated by flow cytometry technique. In addition, the pharmacological cell death inhibitors NEC-1, RIP-1, BAPTA, Z-VAD, and Z-DEVD were used to confirm the related cell death mechanisms. With the results, it was possible to elucidate the novel compound characterized as 2'-OH-Torosaol I. In normal cells, the compound showed no cytotoxicity in PBMC but reduced the cell viability of all melanoma and leukemic cell lines evaluated. 2'-OH-Torosaol I inhibited chemotaxis of B16F10-Nex2, SK-Mel-19, SK-Mel-28 and SK-Mel-103. The cytotoxicity of the compound was induced by apoptosis via the intrinsic pathway with reduced mitochondrial membrane potential, increased levels of reactive oxygen species, and activation of caspase-3. In addition, the inhibitors demonstrated the involvement of necroptosis and Ca2+ in the death process and confirmed caspase-dependent apoptosis death as one of the main programmed cell death pathways induced by 2'-OH-Torosaol I. Taken together, the data characterize the novel anthraquinone 2'-OH-Torosaol I, demonstrating its anticancer activity and potential application in cancer therapy.
Collapse
Affiliation(s)
- David Tsuyoshi Hiramatsu Castro
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| | - Daniel Ferreira Leite
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| | - Debora da Silva Baldivia
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| | - Helder Freitas Dos Santos
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| | - Sikiru Olaitan Balogun
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| | - Denise Brentan da Silva
- Laboratory of Natural Products and Mass Spectrometry, Universidade Federal do Mato Grosso do Sul, Cidade Universitária, Campo Grande 79070-900, Brazil
| | - Carlos Alexandre Carollo
- Laboratory of Natural Products and Mass Spectrometry, Universidade Federal do Mato Grosso do Sul, Cidade Universitária, Campo Grande 79070-900, Brazil
| | - Kely de Picoli Souza
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| | - Edson Lucas Dos Santos
- Research Group on Biotechnology and Bioprospecting Applied to Metabolism (GEBBAM), Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados 79804-970, Brazil
| |
Collapse
|
22
|
Huang Z, Wilson JJ. Structure-Activity Relationships of Metal-Based Inhibitors of the Mitochondrial Calcium Uniporter. ChemMedChem 2023; 18:e202300106. [PMID: 37015871 DOI: 10.1002/cmdc.202300106] [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: 02/23/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/06/2023]
Abstract
The mitochondrial calcium uniporter (MCU) is a transmembrane protein that is responsible for mediating mitochondrial calcium (mCa2+ ) uptake. Given this critical function, the MCU has been implicated as an important target for addressing various human diseases. As such, there has a been growing interest in developing small molecules that can inhibit this protein. To date, metal coordination complexes, particularly multinuclear ruthenium complexes, are the most widely investigated MCU inhibitors due to both their potent inhibitory activities as well as their longstanding use for this application. Recent efforts have expanded the metal-based toolkit for MCU inhibition. This concept paper summarizes the development of new metal-based inhibitors of the MCU and their structure-activity relationships in the context of improving their potential for therapeutic use in managing human diseases related to mCa2+ dysregulation.
Collapse
Affiliation(s)
- Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
23
|
Weiser A, Hermant A, Bermont F, Sizzano F, Karaz S, Alvarez-Illera P, Santo-Domingo J, Sorrentino V, Feige JN, De Marchi U. The mitochondrial calcium uniporter (MCU) activates mitochondrial respiration and enhances mobility by regulating mitochondrial redox state. Redox Biol 2023; 64:102759. [PMID: 37302345 PMCID: PMC10363449 DOI: 10.1016/j.redox.2023.102759] [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: 04/21/2023] [Revised: 05/14/2023] [Accepted: 05/21/2023] [Indexed: 06/13/2023] Open
Abstract
Regulation of mitochondrial redox balance is emerging as a key event for cell signaling in both physiological and pathological conditions. However, the link between the mitochondrial redox state and the modulation of these conditions remains poorly defined. Here, we discovered that activation of the evolutionary conserved mitochondrial calcium uniporter (MCU) modulates mitochondrial redox state. By using mitochondria-targeted redox and calcium sensors and genetic MCU-ablated models, we provide evidence of the causality between MCU activation and net reduction of mitochondrial (but not cytosolic) redox state. Redox modulation of redox-sensitive groups via MCU stimulation is required for maintaining respiratory capacity in primary human myotubes and C. elegans, and boosts mobility in worms. The same benefits are obtained bypassing MCU via direct pharmacological reduction of mitochondrial proteins. Collectively, our results demonstrate that MCU regulates mitochondria redox balance and that this process is required to promote the MCU-dependent effects on mitochondrial respiration and mobility.
Collapse
Affiliation(s)
- Anna Weiser
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland; Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, 85354 Freising, Germany
| | - Aurélie Hermant
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Flavien Bermont
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Federico Sizzano
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Sonia Karaz
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Pilar Alvarez-Illera
- Department of Biochemistry and Molecular Biology, University of Valladolid, Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), 47003 Valladolid, Spain
| | - Jaime Santo-Domingo
- Department of Biochemistry and Molecular Biology, University of Valladolid, Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), 47003 Valladolid, Spain
| | - Vincenzo Sorrentino
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596, Singapore
| | - Jerome N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland
| | - Umberto De Marchi
- Nestlé Institute of Health Sciences, Nestlé Research, EPFL Innovation Park, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
24
|
Dausinas Ni P, Hartman M, Slack J, Basile C, Liu S, Wan J, O'Leary HA. Novel differential calcium regulation of hematopoietic stem and progenitor cells under physiological low oxygen conditions. J Cell Physiol 2023. [PMID: 37051890 DOI: 10.1002/jcp.30942] [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: 03/14/2022] [Revised: 11/28/2022] [Accepted: 12/23/2022] [Indexed: 04/14/2023]
Abstract
Low oxygen bone marrow (BM) niches (~1%-4% low O2 ) provide critical signals for hematopoietic stem/progenitor cells (HSC/HSPCs). Our presented data are the first to investigate live, sorted HSC/HSPCs in their native low O2 conditions. Transcriptional and proteomic analysis uncovered differential Ca2+ regulation that correlated with overlapping phenotypic populations consisting of robust increases of cytosolic and mitochondrial Ca2+ , ABC transporter (ABCG2) expression and sodium/hydrogen exchanger (NHE1) expression in live, HSC/HSPCs remaining in constant low O2. We identified a novel Ca2+ high population in HSPCs predominantly detected in low O2 that displayed enhanced frequency of phenotypic LSK/LSKCD150 in low O2 replating assays compared to Ca2+ low populations. Inhibition of the Ca2+ regulator NHE1 (Cariporide) resulted in attenuation of both the low O2 induced Ca2+ high population and subsequent enhanced maintenance of phenotypic LSK and LSKCD150 during low O2 replating. These data reveal multiple levels of differential Ca2+ regulation in low O2 resulting in phenotypic, signaling, and functional consequences in HSC/HSPCs.
Collapse
Affiliation(s)
- Paige Dausinas Ni
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa Hartman
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jacob Slack
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Heather A O'Leary
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
25
|
Bigham NP, Wilson JJ. Investigation of Cobalt(III) Cage Complexes as Inhibitors of the Mitochondrial Calcium Uniporter. Eur J Inorg Chem 2023; 26:e202200735. [PMID: 37636126 PMCID: PMC10449033 DOI: 10.1002/ejic.202200735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 01/18/2023]
Abstract
The mitochondrial calcium uniporter (MCU) mediates uptake of calcium ions (Ca2+) into the mitochondria, a process that is vital for maintaining normal cellular function. Inhibitors of the MCU, the most promising of which are dinuclear ruthenium coordination compounds, have found use as both therapeutic agents and tools for studying the importance of this ion channel. In this study, six Co3+ cage compounds with sarcophagine-like ligands were assessed for their abilities to inhibit MCU-mediated mitochondrial Ca2+ uptake. These complexes were synthesized and characterized according to literature procedures and then investigated in cellular systems for their MCU-inhibitory activities. Among these six compounds, [Co(sen)]3+ (3, sen = 5-(4-amino-2-azabutyl)-5-methyl-3,7-diaza-1,9-nonanediamine) was identified to be a potent MCU inhibitor, with IC50 values of inhibition of 160 and 180 nM in permeabilized HeLa and HEK293T cells, respectively. Furthermore, the cellular uptake of compound 3 was determined, revealing moderate accumulation in cells. Most notably, 3 was demonstrated to operate in intact cells as an MCU inhibitor. Collectively, this work presents the viability of using cobalt coordination complexes as MCU inhibitors, providing a new direction for researchers to investigate in future studies.
Collapse
Affiliation(s)
- Nicholas P Bigham
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
26
|
Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15041192. [PMID: 36831534 PMCID: PMC9953833 DOI: 10.3390/cancers15041192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer deaths among men in Western countries. Mitochondria, the "powerhouse" of cells, undergo distinctive metabolic and structural dynamics in different types of cancer. PCa cells experience peculiar metabolic changes during their progression from normal epithelial cells to early-stage and, progressively, to late-stage cancer cells. Specifically, healthy cells display a truncated tricarboxylic acid (TCA) cycle and inefficient oxidative phosphorylation (OXPHOS) due to the high accumulation of zinc that impairs the activity of m-aconitase, the enzyme of the TCA cycle responsible for the oxidation of citrate. During the early phase of cancer development, intracellular zinc levels decrease leading to the reactivation of m-aconitase, TCA cycle and OXPHOS. PCa cells change their metabolic features again when progressing to the late stage of cancer. In particular, the Warburg effect was consistently shown to be the main metabolic feature of late-stage PCa cells. However, accumulating evidence sustains that both the TCA cycle and the OXPHOS pathway are still present and active in these cells. The androgen receptor axis as well as mutations in mitochondrial genes involved in metabolic rewiring were shown to play a key role in PCa cell metabolic reprogramming. Mitochondrial structural dynamics, such as biogenesis, fusion/fission and mitophagy, were also observed in PCa cells. In this review, we focus on the mitochondrial metabolic and structural dynamics occurring in PCa during tumor development and progression; their role as effective molecular targets for novel therapeutic strategies in PCa patients is also discussed.
Collapse
|
27
|
Huang Z, MacMillan SN, Wilson JJ. A Fluorogenic Inhibitor of the Mitochondrial Calcium Uniporter. Angew Chem Int Ed Engl 2023; 62:e202214920. [PMID: 36515400 PMCID: PMC9892296 DOI: 10.1002/anie.202214920] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/15/2022]
Abstract
Inhibitors of the mitochondrial calcium uniporter (MCU) are valuable tools for studying the role of mitochondrial Ca2+ in various pathophysiological conditions. In this study, a new fluorogenic MCU inhibitor, RuOCou, is presented. This compound is an analogue of the known MCU inhibitor Ru265 that contains fluorescent axial coumarin carboxylate ligands. Upon aquation of RuOCou and release of the axial coumarin ligands, a simultaneous increase in its MCU-inhibitory activity and fluorescence intensity is observed. The fluorescence response of this compound enabled its aquation to be monitored in both HeLa cell lysates and live HeLa cells. This fluorogenic prodrug represents a potential theranostic MCU inhibitor that can be leveraged for the treatment of human diseases related to MCU activity.
Collapse
Affiliation(s)
- Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
28
|
Chen P, Li Y, Zhou Z, Pan C, Zeng L. Lathyrol promotes ER stress-induced apoptosis and proliferation inhibition in lung cancer cells by targeting SERCA2. Biomed Pharmacother 2023; 158:114123. [PMID: 36521248 DOI: 10.1016/j.biopha.2022.114123] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/26/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Lathyrol is a natural product isolated from the traditional Chinese medicine Semen Euphorbiae with unknown anti-tumor effects. We found that lathyrol had significant inhibitory effect on lung cancer cells by inducing apoptosis and inhibiting proliferation. Subsequently, we demonstrated for the first time that endoplasmic reticulum (ER) stress is a key anti-tumor mechanism of lathyrol. Furthermore, we found that lathyrol can induce ER stress in lung cancer cells by upregulating the protein expression levels of GRP78, PERK, p-eIF2α, CHOP, and ATF4, and the inhibitory effect of lathyrol on lung cancer cells was significantly reversed when cells were pretreated with ER stress inhibitor. In addition, we found that inhibition of SERCA2 resulted in depletion of the ER Ca2+ pool followed by a sustained increase in cytoplasmic Ca2+ levels, eventually leading to ER stress induced tumor cell apoptosis and proliferation inhibition. Lathyrol targeted SERCA2 to cause a significant upregulation of Ca2+ levels, and the inhibitory effect of lathyrol on lung cancer cells was significantly reversed after pretreatment with SERCA2 agonist. Taken together, our data suggest that lathyrol exerts its anti-tumor effect primarily by targeting SERCA2. Our findings highlight the potential for lathyrol as a new candidate drug for the treatment of lung cancer.
Collapse
Affiliation(s)
- Peng Chen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, China; Department of Pharmacy, School of Medicine, Zhejiang University City College, Hangzhou 310015, China.
| | - Yiqian Li
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Zhou Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, China
| | - Chuqi Pan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, China
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, China; Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China.
| |
Collapse
|
29
|
Ma Z, Han H, Zhao Y. Mitochondrial dysfunction-targeted nanosystems for precise tumor therapeutics. Biomaterials 2023; 293:121947. [PMID: 36512861 DOI: 10.1016/j.biomaterials.2022.121947] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria play critical roles in the regulation of the proliferation and apoptosis of cancerous cells. Targeted induction of mitochondrial dysfunction in cancer cells by multifunctional nanosystems for cancer treatment has attracted increasing attention in the past few years. Numerous therapeutic nanosystems have been designed for precise tumor therapy by inducing mitochondrial dysfunction, including reducing adenosine triphosphate, breaking redox homeostasis, inhibiting glycolysis, regulating proteins, membrane potential depolarization, mtDNA damage, mitophagy dysregulation and so on. Understanding the mechanisms of mitochondrial dysfunction would be helpful for efficient treatment of diseases and accelerating the translation of these therapeutic strategies into the clinic. Then, various strategies to construct mitochondria-targeted nanosystems and induce mitochondrial dysfunction are summarized, and the recent research progress regarding precise tumor therapeutics is highlighted. Finally, the major challenges and an outlook in this rapidly developing field are discussed. This review is expected to inspire further development of novel mitochondrial dysfunction-based strategies for precise treatments of cancer and other human diseases.
Collapse
Affiliation(s)
- Zhaoyu Ma
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Heyou Han
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| |
Collapse
|
30
|
Lee W, Song G, Bae H. Glucotropaeolin Promotes Apoptosis by Calcium Dysregulation and Attenuates Cell Migration with FOXM1 Suppression in Pancreatic Cancer Cells. Antioxidants (Basel) 2023; 12:antiox12020257. [PMID: 36829815 PMCID: PMC9952507 DOI: 10.3390/antiox12020257] [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: 12/26/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/25/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has naturally aggressive characteristics including postoperative recurrence, resistance to conventional treatment, and metastasis. Surgical resection with chemotherapeutic agents has been conducted as the major treatment for PDAC. However, surgical treatment is ineffective in the case of advanced cancer, and conventional adjuvant chemotherapy, including gemcitabine and 5-fluorouracil, show low effectiveness due to the high drug resistance of PDAC to this type of treatment. Therefore, the development of innovative therapeutic drugs is crucial to solving the present limitation of conventional drugs. Glucotropaeolin (GT) is a glucosinolate that can be isolated from the Brassicaceae family. GT has exhibited a growth-inhibitory effect against liver and colon cancer cells; however, there is no study regarding the anticancer effect of GT on PDAC. In our study, we determined the antiproliferative effect of GT in PANC-1 and MIA PaCa-2, representative of PDAC. We revealed the intracellular mechanisms underlying the anticancer effect of GT with respect to cell viability, reactive oxygen species (ROS) accumulation, alteration of mitochondrial membrane potential (MMP), calcium dysregulation, cell migration, and the induction of apoptosis. Moreover, GT regulated the signaling pathways related to anticancer in PDAC cells. Finally, the silencing of the forkhead box protein M, a key factor regulating PDAC progression, contributes to the anticancer property of GT in terms of the induction of apoptosis and cell migration. Therefore, GT may be a potential therapeutic drug against PDAC.
Collapse
Affiliation(s)
- Woonghee Lee
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
- Correspondence: (G.S.); (H.B.); Tel.: +82-2-3290-3881 (G.S.); +82-31-201-2686 (H.B.)
| | - Hyocheol Bae
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
- Correspondence: (G.S.); (H.B.); Tel.: +82-2-3290-3881 (G.S.); +82-31-201-2686 (H.B.)
| |
Collapse
|
31
|
Huang Z, Spivey JA, MacMillan SN, Wilson JJ. A ferrocene-containing analogue of the MCU inhibitor Ru265 with increased cell permeability. Inorg Chem Front 2023. [DOI: 10.1039/d2qi02183h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An analogue of the mitochondrial calcium uniporter (MCU) inhibitor Ru265 containing axial ferrocenecarboxylate ligands is reported. This new complex exhibits enhanced cellular uptake compared to the parent compound Ru265.
Collapse
Affiliation(s)
- Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | - Jesse A. Spivey
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | - Samantha N. MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
32
|
Di Gregorio E, Israel S, Staelens M, Tankel G, Shankar K, Tuszyński JA. The distinguishing electrical properties of cancer cells. Phys Life Rev 2022; 43:139-188. [PMID: 36265200 DOI: 10.1016/j.plrev.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
In recent decades, medical research has been primarily focused on the inherited aspect of cancers, despite the reality that only 5-10% of tumours discovered are derived from genetic causes. Cancer is a broad term, and therefore it is inaccurate to address it as a purely genetic disease. Understanding cancer cells' behaviour is the first step in countering them. Behind the scenes, there is a complicated network of environmental factors, DNA errors, metabolic shifts, and electrostatic alterations that build over time and lead to the illness's development. This latter aspect has been analyzed in previous studies, but how the different electrical changes integrate and affect each other is rarely examined. Every cell in the human body possesses electrical properties that are essential for proper behaviour both within and outside of the cell itself. It is not yet clear whether these changes correlate with cell mutation in cancer cells, or only with their subsequent development. Either way, these aspects merit further investigation, especially with regards to their causes and consequences. Trying to block changes at various levels of occurrence or assisting in their prevention could be the key to stopping cells from becoming cancerous. Therefore, a comprehensive understanding of the current knowledge regarding the electrical landscape of cells is much needed. We review four essential electrical characteristics of cells, providing a deep understanding of the electrostatic changes in cancer cells compared to their normal counterparts. In particular, we provide an overview of intracellular and extracellular pH modifications, differences in ionic concentrations in the cytoplasm, transmembrane potential variations, and changes within mitochondria. New therapies targeting or exploiting the electrical properties of cells are developed and tested every year, such as pH-dependent carriers and tumour-treating fields. A brief section regarding the state-of-the-art of these therapies can be found at the end of this review. Finally, we highlight how these alterations integrate and potentially yield indications of cells' malignancy or metastatic index.
Collapse
Affiliation(s)
- Elisabetta Di Gregorio
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Simone Israel
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Michael Staelens
- Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada
| | - Gabriella Tankel
- Department of Mathematics & Statistics, McMaster University, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | - Karthik Shankar
- Department of Electrical & Computer Engineering, University of Alberta, 9211 116 Street NW, Edmonton, T6G 1H9, AB, Canada
| | - Jack A Tuszyński
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada; Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, T6G 1Z2, AB, Canada.
| |
Collapse
|
33
|
Kumar M, Shelly A, Dahiya P, Ray A, Mazumder S. Aeromonas hydrophila inhibits autophagy triggering cytosolic translocation of mtDNA which activates the pro-apoptotic caspase-1/IL-1β-nitric oxide axis in headkidney macrophages. Virulence 2022; 13:60-76. [PMID: 34967692 PMCID: PMC9794009 DOI: 10.1080/21505594.2021.2018767] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanisms underlying Aeromonas hydrophila-pathogenesis are not well understood. Using head kidney macrophages (HKM) of Clarias gariepinus, we previously reported the role of ER-stress in A. hydrophila-induced pathogenesis. Here, we report that PI3K/PLC-induced cytosolic-Ca2+ imbalance induces the expression of pro-apoptotic ER-stress marker, CHOP in A. hydrophila-infected HKM. CHOP promotes HKM apoptosis by inhibiting AKT activation and enhancing JNK signaling. Elevated mitochondrial ROS (mtROS) was recorded which declined significantly by ameliorating ER-stress and in the presence of ER-Ca2+ release modulators (2-APB and dantrolene) and mitochondrial-Ca2+ uptake inhibitor, Ru360, together suggesting the role of ER-mitochondrial Ca2+ dynamics in mtROS generation. Inhibiting mtROS production reduced HKM death implicating the pro-apoptotic role of mtROS in A. hydrophila-pathogenesis. The expression of autophagic proteins (LC3B, beclin-1, and atg 5) was suppressed in the infected HKM. Our results with autophagy-inducer rapamycin demonstrated that impaired autophagy favored the cytosolic accumulation of mitochondrial DNA (mtDNA) and the process depended on mtROS levels. Enhanced caspase-1 activity and IL-1β production was detected and transfection studies coupled with pharmacological inhibitors implicated mtROS/mtDNA axis to be crucial for activating the caspase-1/IL-1β cascade in infected HKM. RNAi studies further suggested the involvement of IL-1β in generating pro-apoptotic NO in A. hydrophila-infected HKM. Our study suggests a novel role of ER-mitochondria cross-talk in regulating A. hydrophila pathogenesis. Based on our observations, we conclude that A. hydrophila induces ER-stress and inhibits mitophagy resulting in mitochondrial dysfunction which leads to mtROS production and translocation of mtDNA into cytosol triggering the activation of caspase-1/IL-1β-mediated NO production, culminating in HKM apoptosis.
Collapse
Affiliation(s)
- Manmohan Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Asha Shelly
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Priyanka Dahiya
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Atish Ray
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India,Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India,CONTACT Shibnath Mazumder Faculty of Life Sciences and Biotechnology
| |
Collapse
|
34
|
Antitumor Activity and Mechanism of Action of the Antimicrobial Peptide AMP-17 on Human Leukemia K562 Cells. Molecules 2022; 27:molecules27228109. [PMID: 36432210 PMCID: PMC9697079 DOI: 10.3390/molecules27228109] [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: 09/21/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
Cancer is one of the most common malignant diseases in the world. Hence, there is an urgent need to search for novel drugs with antitumor activity against cancer cells. AMP-17, a natural antimicrobial peptide derived from Musca domestica, has antimicrobial activity against Gram-positive bacteria, Gram-negative bacteria, and fungi. However, its antitumor activity and potential mechanism of action in cancer cells remain unclear. In this study, we focused on evaluating the in vitro antitumor activity and mechanism of AMP-17 on leukemic K562 cells. The results showed that AMP-17 exhibited anti-proliferative activity on K562 cells with an IC50 value of 58.91 ± 3.57 μg/mL. The membrane integrity of K562 was disrupted and membrane permeability was increased after AMP-17 action. Further observation using SEM and TEM images showed that the cell structure of AMP-17-treated cells was disrupted, with depressions and pore-like breaks on the cell surface, and vacuolated vesicles in the cytoplasm. Furthermore, further mechanistic studies indicated that AMP-17 induced excessive production of reactive oxygen species and calcium ions release in K562 cells, which led to disturbance of mitochondrial membrane potential and blocked ATP synthesis, followed by activation of Caspase-3 to induce apoptosis. In conclusion, these results suggest that the antitumor activity of AMP-17 may be achieved by disrupting cell structure and inducing apoptosis. Therefore, AMP-17 is expected to be a novel potential agent candidate for leukemia treatment.
Collapse
|
35
|
Bigham NP, Huang Z, Spivey J, Woods JJ, MacMillan SN, Wilson JJ. Carboxylate-Capped Analogues of Ru265 Are MCU Inhibitor Prodrugs. Inorg Chem 2022; 61:17299-17312. [PMID: 36260092 PMCID: PMC11649380 DOI: 10.1021/acs.inorgchem.2c02930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The mitochondrial calcium uniporter (MCU) is a transmembrane protein that resides on the inner membrane of the mitochondria and mediates calcium uptake into this organelle. Given the critical role of mitochondrial calcium trafficking in cellular function, inhibitors of this channel have arisen as tools for studying the biological relevance of this process and as potential therapeutic agents. In this study, four new analogues of the previously reported Ru-based MCU inhibitor [ClRu(NH3)4(μ-N)Ru(NH3)4Cl]Cl3 (Ru265) are reported. These compounds, which bear axial carboxylate ligands, are of the general formula [(RCO2)Ru(NH3)4(μ-N)Ru(NH3)4(O2CR)]X3, where X = NO3- or CF3SO3- and R = H (1), CH3 (2), CH2CH3 (3), and (CH2)2CH3 (4). These complexes were fully characterized by IR spectroscopy, NMR spectroscopy, and elemental analysis. X-ray crystal structures of 1 and 3 were obtained, revealing the expected presence of both the linear Ru(μ-N)Ru core and axial formate and propionate ligands. The axial carboxylate ligands of complexes 1-4 are displaced by water in buffered aqueous solution to give the aquated compound Ru265'. The kinetics of these processes were measured by 1H NMR spectroscopy, revealing half-lives that span 5.9-9.9 h at 37 °C. Complex 1 with axial formate ligands underwent aquation approximately twice as fast as the other compounds. In vitro cytotoxicity and mitochondrial membrane potential measurements carried out in HeLa and HEK293T cells demonstrated that none of these four complexes negatively affects cell viability or mitochondrial function. The abilities of 1-4 to inhibit mitochondrial calcium uptake in permeabilized HEK293T cells were assessed and compared to that of Ru265. Fresh solutions of 1-4 are approximately 2-fold less potent than Ru265 with IC50 values in the range of 14.7-19.1 nM. Preincubating 1-4 in aqueous buffers for longer time periods to allow for the aquation reactions to proceed increases their potency of mitochondrial uptake inhibition to match that of Ru265. This result indicates that 1-4 are aquation-activated prodrugs of Ru265'. Finally, 1-4 were shown to inhibit mitochondrial calcium uptake in intact, nonpermeabilized cells, revealing their value as tools and potential therapeutic agents for mitochondrial calcium-related disorders.
Collapse
Affiliation(s)
- Nicholas P Bigham
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jesse Spivey
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Joshua J Woods
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
- Robert F. Smith School of Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
36
|
Uzhachenko R, Shimamoto A, Chirwa SS, Ivanov SV, Ivanova AV, Shanker A. Mitochondrial Fus1/Tusc2 and cellular Ca2 + homeostasis: tumor suppressor, anti-inflammatory and anti-aging implications. Cancer Gene Ther 2022; 29:1307-1320. [PMID: 35181743 PMCID: PMC9576590 DOI: 10.1038/s41417-022-00434-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/22/2021] [Accepted: 01/28/2022] [Indexed: 12/02/2022]
Abstract
FUS1/TUSC2 (FUSion1/TUmor Suppressor Candidate 2) is a tumor suppressor gene (TSG) originally described as a member of the TSG cluster from human 3p21.3 chromosomal region frequently deleted in lung cancer. Its role as a TSG in lung, breast, bone, and other cancers was demonstrated by several groups, but molecular mechanisms of its activities are starting to unveil lately. They suggest that Fus1-dependent mechanisms are relevant in etiologies of diseases beyond cancer, such as chronic inflammation, bacterial and viral infections, premature aging, and geriatric diseases. Here, we revisit the discovery of FUS1 gene in the context of tumor initiation and progression, and review 20 years of research into FUS1 functions and its molecular, structural, and biological aspects that have led to its use in clinical trials and gene therapy. We present a data-driven view on how interactions of Fus1 with the mitochondrial Ca2+ (mitoCa2+) transport machinery maintain cellular Ca2+ homeostasis and control cell apoptosis and senescence. This Fus1-mediated cellular homeostasis is at the crux of tumor suppressor, anti-inflammatory and anti-aging activities.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, USA
| | - Sanika S Chirwa
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Sergey V Ivanov
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Alla V Ivanova
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA.
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA.
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
37
|
Hagiwara H, Watanabe M, Fujioka Y, Kadosaka T, Koizumi T, Koya T, Nakao M, Kamada R, Temma T, Okada K, Moreno JA, Kwon O, Sabe H, Ohba Y, Anzai T. Stimulation of the mitochondrial calcium uniporter mitigates chronic heart failure-associated ventricular arrhythmia in mice. Heart Rhythm 2022; 19:1725-1735. [PMID: 35660475 PMCID: PMC10746330 DOI: 10.1016/j.hrthm.2022.05.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND An aberrant increase in the diastolic calcium concentration ([Ca2+]i) level is a hallmark of heart failure (HF) and the cause of delayed afterdepolarization and ventricular arrhythmia (VA). Although mitochondria play a role in regulating [Ca2+]i, whether they can compensate for the [Ca2+]i abnormality in ventricular myocytes is unknown. OBJECTIVE The purpose of this study was to investigate whether enhanced Ca2+ uptake of mitochondria may compensate for an abnormal increase in the [Ca2+]i of ventricular myocytes in HF to effectively mitigate VA. METHODS We used a HF mouse model in which myocardial infarction was induced by permanent left anterior descending coronary artery ligation. The mitochondrial Ca2+ uniporter was stimulated by kaempferol. Ca2+ dynamics and membrane potential were measured using an epifluorescence microscope, a confocal microscope, and the perforated patch-clamp technique. VA was induced in Langendorff-perfused hearts, and hemodynamic parameters were measured using a microtip transducer catheter. RESULTS Protein expression of the mitochondrial Ca2+ uniporter, as assessed by its subunit expression, did not change between HF and sham mice. Treatment of cardiomyocytes with kaempferol, isolated from HF mice 28 days after coronary ligation, reduced the appearance of aberrant diastolic [Ca2+]i waves and sparks and spontaneous action potentials. Kaempferol effectively reduced VA occurring in Langendorff-perfused hearts. Intravenous administration of kaempferol did not markedly affect left ventricular hemodynamic parameters. CONCLUSION The effects of kaempferol in HF of mice implied that mitochondria may have the potential to compensate for abnormal [Ca2+]i. Mechanisms involved in mitochondrial Ca2+ uptake may provide novel targets for treatment of HF-associated VA.
Collapse
Affiliation(s)
- Hikaru Hagiwara
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaya Watanabe
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahide Kadosaka
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Koizumi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taro Koya
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Motoki Nakao
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rui Kamada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taro Temma
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Jose Antonio Moreno
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California
| | - Ohyun Kwon
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California
| | - Hisakata Sabe
- Department of Molecular Biology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
38
|
Resende R, Fernandes T, Pereira AC, Marques AP, Pereira CF. Endoplasmic Reticulum-Mitochondria Contacts Modulate Reactive Oxygen Species-Mediated Signaling and Oxidative Stress in Brain Disorders: The Key Role of Sigma-1 Receptor. Antioxid Redox Signal 2022; 37:758-780. [PMID: 35369731 DOI: 10.1089/ars.2020.8231] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Mitochondria-Associated Membranes (MAMs) are highly dynamic endoplasmic reticulum (ER)-mitochondria contact sites that, due to the transfer of lipids and Ca2+ between these organelles, modulate several physiologic processes, such as ER stress response, mitochondrial bioenergetics and fission/fusion events, autophagy, and inflammation. In addition, these contacts are implicated in the modulation of the cellular redox status since several MAMs-resident proteins are involved in the generation of reactive oxygen species (ROS), which can act as both signaling mediators and deleterious molecules, depending on their intracellular levels. Recent Advances: In the past few years, structural and functional alterations of MAMs have been associated with the pathophysiology of several neurodegenerative diseases that are closely associated with the impairment of several MAMs-associated events, including perturbation of the redox state on the accumulation of high ROS levels. Critical Issues: Inter-organelle contacts must be tightly regulated to preserve cellular functioning by maintaining Ca2+ and protein homeostasis, lipid metabolism, mitochondrial dynamics and energy production, as well as ROS signaling. Simultaneously, these contacts should avoid mitochondrial Ca2+ overload, which might lead to energetic deficits and deleterious ROS accumulation, culminating in oxidative stress-induced activation of apoptotic cell death pathways, which are common features of many neurodegenerative diseases. Future Directions: Given that Sig-1R is an ER resident chaperone that is highly enriched at the MAMs and that controls ER to mitochondria Ca2+ flux, as well as oxidative and ER stress responses, its potential as a therapeutic target for neurodegenerative diseases such as Amyotrophic Lateral Sclerosis, Alzheimer, Parkinson, and Huntington diseases should be further explored. Antioxid. Redox Signal. 37, 758-780.
Collapse
Affiliation(s)
- Rosa Resende
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Tânia Fernandes
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Catarina Pereira
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Patrícia Marques
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Cláudia Fragão Pereira
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
39
|
Kouba S, Hague F, Ahidouch A, Ouadid-Ahidouch H. Crosstalk between Ca2+ Signaling and Cancer Stemness: The Link to Cisplatin Resistance. Int J Mol Sci 2022; 23:ijms231810687. [PMID: 36142596 PMCID: PMC9503744 DOI: 10.3390/ijms231810687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/07/2022] [Accepted: 09/10/2022] [Indexed: 11/28/2022] Open
Abstract
In the fight against cancer, therapeutic strategies using cisplatin are severely limited by the appearance of a resistant phenotype. While cisplatin is usually efficient at the beginning of the treatment, several patients endure resistance to this agent and face relapse. One of the reasons for this resistant phenotype is the emergence of a cell subpopulation known as cancer stem cells (CSCs). Due to their quiescent phenotype and self-renewal abilities, these cells have recently been recognized as a crucial field of investigation in cancer and treatment resistance. Changes in intracellular calcium (Ca2+) through Ca2+ channel activity are essential for many cellular processes such as proliferation, migration, differentiation, and survival in various cell types. It is now proved that altered Ca2+ signaling is a hallmark of cancer, and several Ca2+ channels have been linked to CSC functions and therapy resistance. Moreover, cisplatin was shown to interfere with Ca2+ homeostasis; thus, it is considered likely that cisplatin-induced aberrant Ca2+ signaling is linked to CSCs biology and, therefore, therapy failure. The molecular signature defining the resistant phenotype varies between tumors, and the number of resistance mechanisms activated in response to a range of pressures dictates the global degree of cisplatin resistance. However, if we can understand the molecular mechanisms linking Ca2+ to cisplatin-induced resistance and CSC behaviors, alternative and novel therapeutic strategies could be considered. In this review, we examine how cisplatin interferes with Ca2+ homeostasis in tumor cells. We also summarize how cisplatin induces CSC markers in cancer. Finally, we highlight the role of Ca2+ in cancer stemness and focus on how they are involved in cisplatin-induced resistance through the increase of cancer stem cell populations and via specific pathways.
Collapse
Affiliation(s)
- Sana Kouba
- Laboratoire de Physiologie Cellulaire et Moléculaire, Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
| | - Frédéric Hague
- Laboratoire de Physiologie Cellulaire et Moléculaire, Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
| | - Ahmed Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire, Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
- Département de Biologie, Faculté des Sciences, Université Ibn Zohr, Agadir 81016, Morocco
| | - Halima Ouadid-Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire, Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
- Correspondence:
| |
Collapse
|
40
|
Jabir NR, Khan MS, Alafaleq NO, Naz H, Ahmed BA. Anticancer potential of yohimbine in drug-resistant oral cancer KB-ChR-8-5 cells. Mol Biol Rep 2022; 49:9565-9573. [PMID: 35970968 DOI: 10.1007/s11033-022-07847-7] [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: 06/07/2022] [Accepted: 08/05/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The demand for environmentally friendly and cost-effective plant-based products for the development of cancer therapeutics has been increasing. Yohimbine (α2-adrenergic receptor antagonist) is a stimulant and aphrodisiac used to improve erectile dysfunction. In this study, we aimed to evaluate the anticancer potential of yohimbine in drug-resistant oral cancer KB-ChR-8-5 cells using different biomolecular techniques. METHODS We estimated the anticancer efficacy of yohimbine using different assays, such as MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cell cytotoxicity, cell morphology, cell apoptosis, reactive oxygen species (ROS) formation, and modulation in the mitochondrial membrane potential (MMP). RESULTS Yohimbine showed a dose-dependent increase in cytotoxicity with a 50% inhibitory concentration (IC50) of 44 µM against KB-ChR-8-5 cancer cell lines. Yohimbine treatment at 40 µM and 50 µM resulted in a considerable change in cell morphology, including shrinkage, detachment, membrane blebbing, and deformed shape. Moreover, at the dose of IC50 and above, a significant induction was observed in the generation of ROS and depolarization of MMP. The possible mechanisms of action of yohimbine underlying the dose-dependent increase in cytotoxicity may be due to the induction of apoptosis, ROS generation, and modulation of MMP. CONCLUSION Overall, yohimbine showed a significant anticancer potential against drug-resistant oral cancer KB-ChR-8-5 cells. Our study suggests that besides being an aphrodisiac, yohimbine can be used as a drug repurposing agent. However, more research is required in different in vitro and in vivo models to confirm the feasibility of yohimbine in clinics.
Collapse
Affiliation(s)
- Nasimudeen R Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, 613403, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Nouf Omar Alafaleq
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Huma Naz
- Department of Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Bakrudeen Ali Ahmed
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, 613403, India.
| |
Collapse
|
41
|
Kumar S, Dhamija B, Attrish D, Sawant V, Sengar M, Thorat J, Shet T, Jain H, Purwar R. Genetic alterations and oxidative stress in T cell lymphomas. Pharmacol Ther 2022; 236:108109. [PMID: 35007658 DOI: 10.1016/j.pharmthera.2022.108109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/20/2022]
Abstract
T cell lymphomas encompass a diverse group of Non-Hodgkin lymphomas with a wide spectrum of clinical, immunological and pathological manifestations. In the last two decades there has been a progress in our understanding of the cell of origin, genetic abnormalities and their impact on behaviour in T cell lymphomas. Genetic alterations are one of the critical drivers of the pathogenesis of T cell lymphoma. Disease progression has been correlated with multiple genetic abnormalities where malignant clones arise primarily out of the host immune surveillance arsenal. There are many cellular processes involved in disease development, and some of them are T cell signaling, differentiation, epigenetic modifications, and immune regulation. Modulation of these crucial pathways via genetic mutations and chromosomal abnormalities possessing either point or copy number mutations helps tumor cells to develop a niche favourable for their growth via metabolic alterations. Several metabolic pathways especially regulation of redox homeostasis is critical in pathogenesis of lymphoma. Disruption of redox potential and induction of oxidative stress renders malignant cells vulnerable to mitochondrial damage and triggers apoptotic pathways causing cell death. Targeting genetic abnormalities and oxidative stress along with current treatment regime have the potential for improved therapeutics and presents new combination approaches towards selective treatment of T cell lymphomas.
Collapse
Affiliation(s)
- Sushant Kumar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Bhavuk Dhamija
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Diksha Attrish
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Vinanti Sawant
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Manju Sengar
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Jayashree Thorat
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Tanuja Shet
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Hasmukh Jain
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Rahul Purwar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India.
| |
Collapse
|
42
|
The Application of the Neuroprotective and Potential Antioxidant Effect of Ergotamine Mediated by Targeting N-Methyl-D-Aspartate Receptors. Antioxidants (Basel) 2022; 11:antiox11081471. [PMID: 36009192 PMCID: PMC9405237 DOI: 10.3390/antiox11081471] [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: 06/22/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: The N-methyl-D-aspartate receptors (NMDARs) mediate fast excitatory currents leading to depolarization. Postsynaptic NMDARs are ionotropic glutamate receptors that mediate excitatory glutamate or glycine signaling in the CNS and play a primary role in long-term potentiation, which is a major form of use-dependent synaptic plasticity. The overstimulation of NMDARs mediates excessive Ca2+ influx to postsynaptic neurons and facilitates more production of ROS, which induces neuronal apoptosis. (2) Methods: To confirm the induced inward currents by the coapplication of glutamate and ergotamine on NMDARs, a two-electrode voltage clamp (TEVC) was conducted. The ergotamine-mediated inhibitory effects of NR1a/NR2A subunits were explored among four different kinds of recombinant NMDA subunits. In silico docking modeling was performed to confirm the main binding site of ergotamine. (3) Results: The ergotamine-mediated inhibitory effect on the NR1a/NR2A subunits has concentration-dependent, reversible, and voltage-independent properties. The major binding sites were V169 of the NR1a subunit and N466 of the NR2A subunit. (4) Conclusion: Ergotamine effectively inhibited NR1a/NR2A subunit among the subtypes of NMDAR. This inhibition effect can prevent excessive Ca2+ influx, which prevents neuronal death.
Collapse
|
43
|
Matairesinol Induces Mitochondrial Dysfunction and Exerts Synergistic Anticancer Effects with 5-Fluorouracil in Pancreatic Cancer Cells. Mar Drugs 2022; 20:md20080473. [PMID: 35892941 PMCID: PMC9331355 DOI: 10.3390/md20080473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer and exhibits a devastating 5-year survival rate. The most recent procedure for the treatment of PDAC is a combination of several conventional chemotherapeutic agents, termed FOLFIRINOX, that includes irinotecan, leucovorin, oxaliplatin, and 5-fluorouracil (5-FU). However, ongoing treatment using these agents is challenging due to their severe side effects and limitations on the range of patients available for PDAC. Therefore, safer and more innovative anticancer agents must be developed. The anticarcinoma activity of matairesinol that can be extracted from seagrass has been reported in various types of cancer, including prostate, breast, cervical, and pancreatic cancer. However, the molecular mechanism of effective anticancer activity of matairesinol against pancreatic cancer remains unclear. In the present study, we confirmed the inhibition of cell proliferation and progression induced by matairesinol in representative human pancreatic cancer cell lines (MIA PaCa-2 and PANC-1). Additionally, matairesinol triggers apoptosis and causes mitochondrial impairment as evidenced by the depolarization of the mitochondrial membrane, disruption of calcium, and suppression of cell migration and related intracellular signaling pathways. Finally, matairesinol exerts a synergistic effect with 5-FU, a standard anticancer agent for PDAC. These results demonstrate the therapeutic potential of matairesinol in the treatment of PDAC.
Collapse
|
44
|
Suárez-Rivero JM, Pastor-Maldonado CJ, Povea-Cabello S, Álvarez-Córdoba M, Villalón-García I, Talaverón-Rey M, Suárez-Carrillo A, Munuera-Cabeza M, Reche-López D, Cilleros-Holgado P, Piñero-Pérez R, Sánchez-Alcázar JA. Activation of the Mitochondrial Unfolded Protein Response: A New Therapeutic Target? Biomedicines 2022; 10:1611. [PMID: 35884915 PMCID: PMC9313171 DOI: 10.3390/biomedicines10071611] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction is a key hub that is common to many diseases. Mitochondria's role in energy production, calcium homeostasis, and ROS balance makes them essential for cell survival and fitness. However, there are no effective treatments for most mitochondrial and related diseases to this day. Therefore, new therapeutic approaches, such as activation of the mitochondrial unfolded protein response (UPRmt), are being examined. UPRmt englobes several compensation processes related to proteostasis and antioxidant mechanisms. UPRmt activation, through an hormetic response, promotes cell homeostasis and improves lifespan and disease conditions in biological models of neurodegenerative diseases, cardiopathies, and mitochondrial diseases. Although UPRmt activation is a promising therapeutic option for many conditions, its overactivation could lead to non-desired side effects, such as increased heteroplasmy of mitochondrial DNA mutations or cancer progression in oncologic patients. In this review, we present the most recent UPRmt activation therapeutic strategies, UPRmt's role in diseases, and its possible negative consequences in particular pathological conditions.
Collapse
Affiliation(s)
- Juan M. Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Carmen J. Pastor-Maldonado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
| | - José A. Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013 Sevilla, Spain; (J.M.S.-R.); (C.J.P.-M.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.R.-L.); (P.C.-H.); (R.P.-P.)
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013 Sevilla, Spain
| |
Collapse
|
45
|
Wang X, Li Y, Li Z, Lin S, Wang H, Sun J, Lan C, Wu L, Sun D, Huang C, Singh PK, Hempel N, Trebak M, DeNicola GM, Hao J, Yang S. Mitochondrial Calcium Uniporter Drives Metastasis and Confers a Targetable Cystine Dependency in Pancreatic Cancer. Cancer Res 2022; 82:2254-2268. [PMID: 35413105 PMCID: PMC9203979 DOI: 10.1158/0008-5472.can-21-3230] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/01/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease with few effective treatments. Here we show that the mitochondrial calcium uniporter (MCU) promotes PDAC cell migration, invasion, metastasis, and metabolic stress resistance by activating the Keap1-Nrf2 antioxidant program. The cystine transporter SLC7A11 was identified as a druggable target downstream of the MCU-Nrf2 axis. Paradoxically, despite the increased ability to uptake cystine, MCU-overexpressing PDAC demonstrated characteristics typical of cystine-deprived cells and were hypersensitive to cystine deprivation-induced ferroptosis. Pharmacologic inhibitors of SLC7A11 effectively induced tumor regression and abrogated MCU-driven metastasis in PDAC. In patient-derived organoid models in vitro and patient-derived xenograft models in vivo, MCU-high PDAC demonstrated increased sensitivity to SLC7A11 inhibition compared with MCU-low tumors. These data suggest that MCU is able to promote resistance to metabolic stress and to drive PDAC metastasis in a cystine-dependent manner. MCU-mediated cystine addiction could be exploited as a therapeutic vulnerability to inhibit PDAC tumor growth and to prevent metastasis. SIGNIFICANCE Elevated mitochondrial calcium uptake in PDAC promotes metastasis but exposes cystine addiction and ferroptosis sensitivity that could be targeted to improve pancreatic cancer treatment.
Collapse
Affiliation(s)
- Xiuchao Wang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yunzhan Li
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Zekun Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Shengchen Lin
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Hongwei Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jianwei Sun
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life-Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Chungen Lan
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Liangliang Wu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Dongxiao Sun
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chongbiao Huang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Pankaj K. Singh
- Eppley Institute for Research in Cancer and Allied Diseases, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Nadine Hempel
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Gina M. DeNicola
- Department of Cancer Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
46
|
Yang Y, Zhao Y, Zhang Y, Niu L, Li W, Lu W, Li J, Schäfer P, Meng Y, Shan W. A mitochondrial RNA processing protein mediates plant immunity to a broad spectrum of pathogens by modulating the mitochondrial oxidative burst. THE PLANT CELL 2022; 34:2343-2363. [PMID: 35262740 PMCID: PMC9134091 DOI: 10.1093/plcell/koac082] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/18/2022] [Indexed: 06/01/2023]
Abstract
Mitochondrial function depends on the RNA processing of mitochondrial gene transcripts by nucleus-encoded proteins. This posttranscriptional processing involves the large group of nuclear-encoded pentatricopeptide repeat (PPR) proteins. Mitochondrial processes represent a crucial part in animal immunity, but whether mitochondria play similar roles in plants remains unclear. Here, we report the identification of RESISTANCE TO PHYTOPHTHORA PARASITICA 7 (AtRTP7), a P-type PPR protein, in Arabidopsis thaliana and its conserved function in immunity to diverse pathogens across distantly related plant species. RTP7 affects the levels of mitochondrial reactive oxygen species (mROS) by participating in RNA splicing of nad7, which encodes a critical subunit of the mitochondrial respiratory chain Complex I, the largest of the four major components of the mitochondrial oxidative phosphorylation system. The enhanced resistance of rtp7 plants to Phytophthora parasitica is dependent on an elevated mROS burst, but might be independent from the ROS burst associated with plasma membrane-localized NADPH oxidases. Our study reveals the immune function of RTP7 and the defective processing of Complex I subunits in rtp7 plants resulted in enhanced resistance to both biotrophic and necrotrophic pathogens without affecting overall plant development.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Agronomy, Northwest A&F University, Yangling 712100, China
| | - Yan Zhao
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Agronomy, Northwest A&F University, Yangling 712100, China
| | - Yingqi Zhang
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Agronomy, Northwest A&F University, Yangling 712100, China
| | - Lihua Niu
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Agronomy, Northwest A&F University, Yangling 712100, China
| | - Wanyue Li
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Agronomy, Northwest A&F University, Yangling 712100, China
| | - Wenqin Lu
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Plant Protection, Northwest A&F University, Yangling 712100, China
| | - Jinfang Li
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Plant Protection, Northwest A&F University, Yangling 712100, China
| | - Patrick Schäfer
- Institute of Molecular Botany, Ulm University, Ulm 89069, Germany
| | - Yuling Meng
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Agronomy, Northwest A&F University, Yangling 712100, China
| | | |
Collapse
|
47
|
Shyam R, Ogando DG, Bonanno JA. Mitochondrial ROS in Slc4a11 KO Corneal Endothelial Cells Lead to ER Stress. Front Cell Dev Biol 2022; 10:878395. [PMID: 35557943 PMCID: PMC9086159 DOI: 10.3389/fcell.2022.878395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Recent studies from Slc4a11 -/- mice have identified glutamine-induced mitochondrial dysfunction as a significant contributor toward oxidative stress, impaired lysosomal function, aberrant autophagy, and cell death in this Congenital Hereditary Endothelial Dystrophy (CHED) model. Because lysosomes are derived from endoplasmic reticulum (ER)-Golgi, we asked whether ER function is affected by mitochondrial ROS in Slc4a11 KO corneal endothelial cells. In mouse Slc4a11 -/- corneal endothelial tissue, we observed the presence of dilated ER and elevated expression of ER stress markers BIP and CHOP. Slc4a11 KO mouse corneal endothelial cells incubated with glutamine showed increased aggresome formation, BIP and GADD153, as well as reduced ER Ca2+ release as compared to WT. Induction of mitoROS by ETC inhibition also led to ER stress in WT cells. Treatment with the mitochondrial ROS quencher MitoQ, restored ER Ca2+ release and relieved ER stress markers in Slc4a11 KO cells in vitro. Systemic MitoQ also reduced BIP expression in Slc4a11 KO endothelium. We conclude that mitochondrial ROS can induce ER stress in corneal endothelial cells.
Collapse
Affiliation(s)
- Rajalekshmy Shyam
- Vision Science Program, School of Optometry, Indiana University, Bloomington, IN, United States
| | | | | |
Collapse
|
48
|
Chiu HY, Loh AHP, Taneja R. Mitochondrial calcium uptake regulates tumour progression in embryonal rhabdomyosarcoma. Cell Death Dis 2022; 13:419. [PMID: 35490194 PMCID: PMC9056521 DOI: 10.1038/s41419-022-04835-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/09/2022]
Abstract
AbstractEmbryonal rhabdomyosarcoma (ERMS) is characterised by a failure of cells to complete skeletal muscle differentiation. Although ERMS cells are vulnerable to oxidative stress, the relevance of mitochondrial calcium homoeostasis in oncogenesis is unclear. Here, we show that ERMS cell lines as well as primary tumours exhibit elevated expression of the mitochondrial calcium uniporter (MCU). MCU knockdown resulted in impaired mitochondrial calcium uptake and a reduction in mitochondrial reactive oxygen species (mROS) levels. Phenotypically, MCU knockdown cells exhibited reduced cellular proliferation and motility, with an increased propensity to differentiate in vitro and in vivo. RNA-sequencing of MCU knockdown cells revealed a significant reduction in genes involved in TGFβ signalling that play prominent roles in oncogenesis and inhibition of myogenic differentiation. Interestingly, modulation of mROS production impacted TGFβ signalling. Our study elucidates mechanisms by which mitochondrial calcium dysregulation promotes tumour progression and suggests that targeting the MCU complex to restore mitochondrial calcium homoeostasis could be a therapeutic avenue in ERMS.
Collapse
|
49
|
Lon upregulation contributes to cisplatin resistance by triggering NCLX-mediated mitochondrial Ca 2+ release in cancer cells. Cell Death Dis 2022; 13:241. [PMID: 35296653 PMCID: PMC8927349 DOI: 10.1038/s41419-022-04668-1] [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: 07/23/2021] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022]
Abstract
Mitochondria are the major organelles in sensing cellular stress and inducing the response for cell survival. Mitochondrial Lon has been identified as an important stress protein involved in regulating proliferation, metastasis, and apoptosis in cancer cells. However, the mechanism of retrograde signaling by Lon on mitochondrial DNA (mtDNA) damage remains to be elucidated. Here we report the role of Lon in the response to cisplatin-induced mtDNA damage and oxidative stress, which confers cancer cells on cisplatin resistance via modulating calcium levels in mitochondria and cytosol. First, we found that cisplatin treatment on oral cancer cells caused oxidative damage of mtDNA and induced Lon expression. Lon overexpression in cancer cells decreased while Lon knockdown sensitized the cytotoxicity towards cisplatin treatment. We further identified that cisplatin-induced Lon activates the PYK2-SRC-STAT3 pathway to stimulate Bcl-2 and IL-6 expression, leading to the cytotoxicity resistance to cisplatin. Intriguingly, we found that activation of this pathway is through an increase of intracellular calcium (Ca2+) via NCLX, a mitochondrial Na+/Ca2+ exchanger. We then verified that NCLX expression is dependent on Lon levels; Lon interacts with and activates NCLX activity. NCLX inhibition increased the level of mitochondrial calcium and sensitized the cytotoxicity to cisplatin in vitro and in vivo. In summary, mitochondrial Lon-induced cisplatin resistance is mediated by calcium release into cytosol through NCLX, which activates calcium-dependent PYK2-SRC-STAT3-IL-6 pathway. Thus, our work uncovers the novel retrograde signaling by mitochondrial Lon on resistance to cisplatin-induced mtDNA stress, indicating the potential use of Lon and NCLX inhibitors for better clinical outcomes in chemoresistant cancer patients.
Collapse
|
50
|
Khan MA, Singh R, Siddiqui S, Ahmad I, Ahmad R, Upadhyay S, Barkat MA, Ali AMA, Zia Q, Srivastava A, Trivedi A, Husain I, Srivastava AN, Mishra DP. Anticancer potential of Phoenix dactylifera L. seed extract in human cancer cells and pro-apoptotic effects mediated through caspase-3 dependent pathway in human breast cancer MDA-MB-231 cells: an in vitro and in silico investigation. BMC Complement Med Ther 2022; 22:68. [PMID: 35291987 PMCID: PMC8922853 DOI: 10.1186/s12906-022-03533-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 02/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background Phoenix dactylifera L. has a diverse set of pharmacological properties due to its distinct phytochemical profile. The purpose of this study was to investigate the anticancer potential of Phoenix dactylifera seed extract (PDSE) in human breast cancer MDA-MB-231 and MCF-7 cells, as well as liver cancer HepG2 cells, and to investigate the anticancer efficacy in triple-negative MDA-MB-231 cells, followed by in silico validation of the molecular interaction between active components of PDSE and caspase-3, an apoptosis executioner protein . Methods In this study, human cancer cell lines were cultured and subsequently treated with 10 to 100 μg/mL of PDSE. MTT test was performed to determine the cell viability, MMP was measured using fluorescent probe JC-1, nuclear condensation was determined by Hoechst 33258 dye, Annexin V-FITC & PI staining and cell cycle analysis were evaluated through flow cytometer, and apoptotic markers were detected using western blotting. The bioactive agents in PDSE were identified using high-performance liquid chromatography (HPLC) analysis. The binding affinity was validated using molecular docking tools AutoDock Vina and iGEMDOCK v2.1. Results Cell viability data indicated that PDSE inhibited cell proliferation in both breast cancer cells and liver cancer cells. MDA-MB-231 cells showed maximum growth inhibition with an IC50 value of 85.86 μg/mL for PDSE. However, PDSE did not show any significant toxicity against the normal Vero cell line. PDSE induced MMP loss and formation of apoptotic bodies, enhanced late apoptosis at high doses and arrested cells in the S phase of cell cycle. PDSE activated the enzymatic activity of cleaved caspase-3 and caused the cleavage of poly-ADB ribose polymerase (PARP) protein. PDSE upregulated pro-apoptotic Bax protein markedly but no significant effect on tumor suppressor protein p53, while it downregulated the anti-apoptotic Bcl-2 protein expression. HPLC analysis showed the presence of rutin and quercetin bioactive flavonols in ethanolic extract of PDS. Interestingly, both active components revealed a strong binding interaction with amino acid residues of caspase-3 (PDB ID: 2XYP; Hetero 4-mer - A2B2) protein. Conclusion PDS could serve as a potential medicinal source for apoptotic cell death in human breast cancer cells and, thus, could be used as a promising and crucial candidate in anticancer drug development. This study warrants further in vivo research, followed by clinical investigation. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03533-0.
Collapse
Affiliation(s)
- Mohsin Ali Khan
- Research and Development Unit, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Romila Singh
- Cell Death Research Laboratory, LSS-106, Endocrinology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Sahabjada Siddiqui
- Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India.
| | - Imran Ahmad
- Department of Biochemistry, King George's Medical University, Lucknow, 226003, India
| | - Rumana Ahmad
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Shivbrat Upadhyay
- Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Ahmed Mahmoud Abdelhaleem Ali
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P. O. Box 11099, Taif, 21944, Saudi Arabia
| | - Qamar Zia
- Health and Basic Science Research Centre, Majmaah University, Majmaah, 11952, Saudi Arabia.,Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, 11952, Saudi Arabia
| | - Aditi Srivastava
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Anchal Trivedi
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Ishrat Husain
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Anand Narain Srivastava
- Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, 226003, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, LSS-106, Endocrinology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India.
| |
Collapse
|