1
|
El Gaafary M, Abdel-Baki PM, El-Halawany AM, Mohamed HM, Duweb A, Abdallah HM, Mohamed GA, Ibrahim SRM, Simmet T, Syrovets T. Prenylated xanthones from mangosteen (Garcinia mangostana) target oxidative mitochondrial respiration in cancer cells. Biomed Pharmacother 2024; 179:117365. [PMID: 39217837 DOI: 10.1016/j.biopha.2024.117365] [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: 06/10/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Mangosteen (Garcinia mangostana) is well-known for its nutritional value and health benefits. Breast cancer is the most common cancer and the leading cause of cancer-related mortality among females worldwide. Here we show that the prenylated xanthones, α-mangostin, γ-mangostin, 9-hydroxycalabaxanthone (9-HCX), and garcinone E from the mangosteen pericarp exhibit cytotoxicity against a panel of human cancer cell lines including lung adenocarcinoma (A549), cervical carcinoma (HeLa), prostatic carcinoma (DU 145), pancreatic carcinoma (MIA PaCa-2), hepatocellular carcinoma (Hep G2), bladder urothelial cancer (5637), as well as the triple-negative breast cancer cells MDA-MB-231. In line with its higher predicted bioactivity score compared to other prenylated xanthones, 9-HCX induced the strongest antiproliferative and proapoptotic effects in MDA-MB-231 breast cancer xenografts in vivo. In different in vitro models, we demonstrate that prenylated xanthones from G. mangostana target mitochondria in cancer cells by inhibition of the mitochondrial respiratory chain complex II (α-mangostin, γ-mangostin, and garcinone E) and complex III (9-HCX) as shown in isolated mitochondria. Accordingly, oxidative mitochondrial respiration (OXPHOS) was inhibited, mitochondrial proton leak increased, and adenosine triphosphate (ATP) synthesis decreased as analyzed by Seahorse assay in MDA-MB-231 cells. Hence, the prenylated xanthones increased mitochondrial superoxide levels, induced mitochondrial membrane permeabilization, and initiated caspase 3/7-mediated apoptosis in MDA-MB-231 triple-negative breast cancer cells. Thus, prenylated xanthones from Garcinia mangostana exhibit anticancer activity based on interference with the mitochondrial respiration.
Collapse
Affiliation(s)
- Menna El Gaafary
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany; Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Passent M Abdel-Baki
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Ali M El-Halawany
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Heba M Mohamed
- Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; Faculty of Health Sciences, Higher Colleges of Technology, Dubai, United Arab Emirates.
| | - Amira Duweb
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany; Department of Pharmacology, Faculty of Medicine, University of Tripoli, Tripoli, Libya.
| | - Hossam M Abdallah
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany; Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Sabrin R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah 21442, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Thomas Simmet
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany.
| | - Tatiana Syrovets
- Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University, D-89081 Ulm, Germany.
| |
Collapse
|
2
|
Luo X, Hu C, Yin Q, Zhang X, Liu Z, Zhou C, Zhang J, Chen W, Yang Y. Dual-Mechanism Peptide SR25 has Broad Antimicrobial Activity and Potential Application for Healing Bacteria-infected Diabetic Wounds. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401793. [PMID: 38874469 PMCID: PMC11321617 DOI: 10.1002/advs.202401793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/12/2024] [Indexed: 06/15/2024]
Abstract
The rise of antibiotic resistance poses a significant public health crisis, particularly due to limited antimicrobial options for the treatment of infections with Gram-negative pathogens. Here, an antimicrobial peptide (AMP) SR25 is characterized, which effectively kills both Gram-negative and Gram-positive bacteria through a unique dual-targeting mechanism without detectable resistance. Meanwhile, an SR25-functionalized hydrogel is developed for the efficient treatment of infected diabetic wounds. SR25 is obtained through genome mining from an uncultured bovine enteric actinomycete named Nonomuraea Jilinensis sp. nov. Investigations reveal that SR25 has two independent cellular targets, disrupting bacterial membrane integrity and restraining the activity of succinate:quinone oxidoreductase (SQR). In a diabetic mice wound infection model, the SR25-incorporated hydrogel exhibits high efficacy against mixed infections of Escherichia coli (E. coli) and methicillin-resistant Staphylococcus aureus (MRSA), accelerating wound healing. Overall, these findings demonstrate the therapeutic potential of SR25 and highlight the value of mining drugs with multiple mechanisms from uncultured animal commensals for combating challenging bacterial pathogens.
Collapse
Affiliation(s)
- Xue‐Yue Luo
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Chun‐Mei Hu
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Qi Yin
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Xiao‐Mei Zhang
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Zhen‐Zhen Liu
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Cheng‐Kai Zhou
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Jian‐Gang Zhang
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Wei Chen
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| | - Yong‐Jun Yang
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineJilin UniversityChangchunJilin130062P. R. China
| |
Collapse
|
3
|
Kumar M, Sharma S, Kumar J, Barik S, Mazumder S. Mitochondrial electron transport chain in macrophage reprogramming: Potential role in antibacterial immune response. CURRENT RESEARCH IN IMMUNOLOGY 2024; 5:100077. [PMID: 38572399 PMCID: PMC10987323 DOI: 10.1016/j.crimmu.2024.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Macrophages restrain microbial infection and reinstate tissue homeostasis. The mitochondria govern macrophage metabolism and serve as pivot in innate immunity, thus acting as immunometabolic regulon. Metabolic pathways produce electron flows that end up in mitochondrial electron transport chain (mtETC), made of super-complexes regulating multitude of molecular and biochemical processes. Cell-intrinsic and extrinsic factors influence mtETC structure and function, impacting several aspects of macrophage immunity. These factors provide the macrophages with alternate fuel sources and metabolites, critical to gain functional competence and overcoming pathogenic stress. Mitochondrial reactive oxygen species (mtROS) and oxidative phosphorylation (OXPHOS) generated through the mtETC are important innate immune attributes, which help macrophages in mounting antibacterial responses. Recent studies have demonstrated the role of mtETC in governing mitochondrial dynamics and macrophage polarization (M1/M2). M1 macrophages are important for containing bacterial pathogens and M2 macrophages promote tissue repair and wound healing. Thus, mitochondrial bioenergetics and metabolism are intimately coupled with innate immunity. In this review, we have addressed mtETC function as innate rheostats that regulate macrophage reprogramming and innate immune responses. Advancement in this field encourages further exploration and provides potential novel macrophage-based therapeutic targets to control unsolicited inflammation.
Collapse
Affiliation(s)
- Manmohan Kumar
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Zoology, Gargi College, University of Delhi, Delhi, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Sailen Barik
- EonBio, 3780 Pelham Drive, Mobile, AL 36619, USA
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, India
| |
Collapse
|
4
|
Sharma P, Maklashina E, Voehler M, Balintova S, Dvorakova S, Kraus M, Hadrava Vanova K, Nahacka Z, Zobalova R, Boukalova S, Cunatova K, Mracek T, Ghayee HK, Pacak K, Rohlena J, Neuzil J, Cecchini G, Iverson TM. Disordered-to-ordered transitions in assembly factors allow the complex II catalytic subunit to switch binding partners. Nat Commun 2024; 15:473. [PMID: 38212624 PMCID: PMC10784507 DOI: 10.1038/s41467-023-44563-7] [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: 11/29/2022] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
Complex II (CII) activity controls phenomena that require crosstalk between metabolism and signaling, including neurodegeneration, cancer metabolism, immune activation, and ischemia-reperfusion injury. CII activity can be regulated at the level of assembly, a process that leverages metastable assembly intermediates. The nature of these intermediates and how CII subunits transfer between metastable complexes remains unclear. In this work, we identify metastable species containing the SDHA subunit and its assembly factors, and we assign a preferred temporal sequence of appearance of these species during CII assembly. Structures of two species show that the assembly factors undergo disordered-to-ordered transitions without the appearance of significant secondary structure. The findings identify that intrinsically disordered regions are critical in regulating CII assembly, an observation that has implications for the control of assembly in other biomolecular complexes.
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Elena Maklashina
- Molecular Biology Division, San Francisco VA Health Care System, San Francisco, CA, 94121, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, 94158, USA
| | - Markus Voehler
- Department of Chemistry Vanderbilt University, Nashville, TN, 37232, USA
- Center for Structural Biology Vanderbilt University, Nashville, TN, 37232, USA
| | - Sona Balintova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic
| | - Sarka Dvorakova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Michal Kraus
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Katerina Hadrava Vanova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Stepana Boukalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Kristyna Cunatova
- Institute of Physiology, Czech Academy of Sciences, Prague 4, 142 20, Prague, Czech Republic
| | - Tomas Mracek
- Institute of Physiology, Czech Academy of Sciences, Prague 4, 142 20, Prague, Czech Republic
| | - Hans K Ghayee
- Department of Medicine, Division of Endocrinology & Metabolism, University of Florida College of Medicine and Malcom Randall, VA Medical Center, Gainesville, FL, 32608, USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic.
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic.
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, 4222, Australia.
- 1st Faculty of Medicine, Charles University, 128 00, Prague 2, Czech Republic.
| | - Gary Cecchini
- Molecular Biology Division, San Francisco VA Health Care System, San Francisco, CA, 94121, USA.
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, 94158, USA.
| | - T M Iverson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Center for Structural Biology Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
5
|
Grayson C, Mailloux RJ. Coenzyme Q 10 and nicotinamide nucleotide transhydrogenase: Sentinels for mitochondrial hydrogen peroxide signaling. Free Radic Biol Med 2023; 208:260-271. [PMID: 37573896 DOI: 10.1016/j.freeradbiomed.2023.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/21/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Mitochondria use hydrogen peroxide (H2O2) as a mitokine for cell communication. H2O2 output for signaling depends on its rate of production and degradation, both of which are strongly affected by the redox state of the coenzyme Q10 (CoQ) pool and NADPH availability. Here, we propose the CoQ pool and nicotinamide nucleotide transhydrogenase (NNT) have evolved to be central modalities for mitochondrial H2O2 signaling. Both factors play opposing yet equally important roles in dictating H2O2 availability because they are connected to one another by two central parameters in bioenergetics: electron supply and Δp. The CoQ pool is the central point of convergence for electrons from various dehydrogenases and the electron transport chain (ETC). The increase in Δp creates a significant amount of protonic backpressure on mitochondria to promote H2O2 genesis through CoQ pool reduction. These same factors also drive the activity of NNT, which uses electrons and the Δp to eliminate H2O2. In this way, electron supply and the magnitude of the Δp manifests as a redox connection between the two sentinels, CoQ and NNT, which serve as opposing yet equally important forces required for budgeting H2O2. Taken together, CoQ and NNT are sentinels linked through mitochondrial bioenergetics to manage H2O2 availability for interorganelle and intercellular redox signaling.
Collapse
Affiliation(s)
- Cathryn Grayson
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
6
|
Duarte Hospital C, Tête A, Debizet K, Imler J, Tomkiewicz-Raulet C, Blanc EB, Barouki R, Coumoul X, Bortoli S. SDHi fungicides: An example of mitotoxic pesticides targeting the succinate dehydrogenase complex. ENVIRONMENT INTERNATIONAL 2023; 180:108219. [PMID: 37778286 DOI: 10.1016/j.envint.2023.108219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Succinate dehydrogenase inhibitors (SDHi) are fungicides used to control the proliferation of pathogenic fungi in crops. Their mode of action is based on blocking the activity of succinate dehydrogenase (SDH), a universal enzyme expressed by all species harboring mitochondria. The SDH is involved in two interconnected metabolic processes for energy production: the transfer of electrons in the mitochondrial respiratory chain and the oxidation of succinate to fumarate in the Krebs cycle. In humans, inherited SDH deficiencies may cause major pathologies including encephalopathies and cancers. The cellular and molecular mechanisms related to such genetic inactivation have been well described in neuroendocrine tumors, in which it induces an oxidative stress, a pseudohypoxic phenotype, a metabolic, epigenetic and transcriptomic remodeling, and alterations in the migration and invasion capacities of cancer cells, in connection with the accumulation of succinate, an oncometabolite, substrate of the SDH. We will discuss recent studies reporting toxic effects of SDHi in non-target organisms and their implications for risk assessment of pesticides. Recent data show that the SDH structure is highly conserved during evolution and that SDHi can inhibit SDH activity in mitochondria of non-target species, including humans. These observations suggest that SDHi are not specific inhibitors of fungal SDH. We hypothesize that SDHi could have toxic effects in other species, including humans. Moreover, the analysis of regulatory assessment reports shows that most SDHi induce tumors in animals without evidence of genotoxicity. Thus, these substances could have a non-genotoxic mechanism of carcinogenicity that still needs to be fully characterized and that could be related to SDH inhibition. The use of pesticides targeting mitochondrial enzymes encoded by tumor suppressor genes raises questions on the risk assessment framework of mitotoxic pesticides. The issue of SDHi fungicides is therefore a textbook case that highlights the urgent need for changes in regulatory assessment.
Collapse
Affiliation(s)
| | - Arnaud Tête
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Kloé Debizet
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Jules Imler
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | | | - Etienne B Blanc
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Robert Barouki
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Xavier Coumoul
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris.
| | - Sylvie Bortoli
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris.
| |
Collapse
|
7
|
Iverson TM, Singh PK, Cecchini G. An evolving view of complex II-noncanonical complexes, megacomplexes, respiration, signaling, and beyond. J Biol Chem 2023; 299:104761. [PMID: 37119852 PMCID: PMC10238741 DOI: 10.1016/j.jbc.2023.104761] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023] Open
Abstract
Mitochondrial complex II is traditionally studied for its participation in two key respiratory processes: the electron transport chain and the Krebs cycle. There is now a rich body of literature explaining how complex II contributes to respiration. However, more recent research shows that not all of the pathologies associated with altered complex II activity clearly correlate with this respiratory role. Complex II activity has now been shown to be necessary for a range of biological processes peripherally related to respiration, including metabolic control, inflammation, and cell fate. Integration of findings from multiple types of studies suggests that complex II both participates in respiration and controls multiple succinate-dependent signal transduction pathways. Thus, the emerging view is that the true biological function of complex II is well beyond respiration. This review uses a semichronological approach to highlight major paradigm shifts that occurred over time. Special emphasis is given to the more recently identified functions of complex II and its subunits because these findings have infused new directions into an established field.
Collapse
Affiliation(s)
- T M Iverson
- Departments of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Departments of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.
| | - Prashant K Singh
- Departments of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Gary Cecchini
- Molecular Biology Division, San Francisco VA Health Care System, San Francisco, California, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, California, USA.
| |
Collapse
|
8
|
Wang Q, Li M, Zeng N, Zhou Y, Yan J. Succinate dehydrogenase complex subunit C: Role in cellular physiology and disease. Exp Biol Med (Maywood) 2023; 248:263-270. [PMID: 36691338 PMCID: PMC10107392 DOI: 10.1177/15353702221147567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Succinate dehydrogenase complex subunit C (SDHC) is a subunit of mitochondrial complex II (MCII), which is also known as succinate dehydrogenase (SDH) or succinate: ubiquinone oxidoreductase. Mitochondrial complex II is the smallest respiratory complex in the respiratory chain and contains four subunits. SDHC is a membrane-anchored subunit of SDH, which connects the tricarboxylic acid cycle and the electron transport chain. SDH regulates several physiological processes within cells, plays an important role in generating energy to maintain normal cell growth, and is involved in apoptosis. Currently, SDHC is generally recognized as a tumor-suppressor gene. SDHC mutations can cause oxidative damage in the body. It is closely related to the occurrence and development of cancer, neurodegenerative diseases, and aging-related diseases. Here, we review studies on the structure, biological function, related diseases of SDHC, and the mev-1 Animal Model of SDHC Mutation and its potential use as a therapeutic target of certain human diseases.
Collapse
Affiliation(s)
- Qi Wang
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Mao Li
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Nannan Zeng
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| | - Yali Zhou
- Department of Microbiology, Guilin Medical University, Guilin 541004, China
| | - Jianguo Yan
- Department of Physiology, Guilin Medical University, Guilin 541004, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541004, China
| |
Collapse
|
9
|
Blood leukocyte transcriptional modules and differentially expressed genes associated with disease severity and age in COVID-19 patients. Sci Rep 2023; 13:898. [PMID: 36650374 PMCID: PMC9844197 DOI: 10.1038/s41598-023-28227-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
Since the molecular mechanisms determining COVID-19 severity are not yet well understood, there is a demand for biomarkers derived from comparative transcriptome analyses of mild and severe cases, combined with patients' clinico-demographic and laboratory data. Here the transcriptomic response of human leukocytes to SARS-CoV-2 infection was investigated by focusing on the differences between mild and severe cases and between age subgroups (younger and older adults). Three transcriptional modules correlated with these traits were functionally characterized, as well as 23 differentially expressed genes (DEGs) associated to disease severity. One module, correlated with severe cases and older patients, had an overrepresentation of genes involved in innate immune response and in neutrophil activation, whereas two other modules, correlated with disease severity and younger patients, harbored genes involved in the innate immune response to viral infections, and in the regulation of this response. This transcriptomic mechanism could be related to the better outcome observed in younger COVID-19 patients. The DEGs, all hyper-expressed in the group of severe cases, were mostly involved in neutrophil activation and in the p53 pathway, therefore related to inflammation and lymphopenia. These biomarkers may be useful for getting a better stratification of risk factors in COVID-19.
Collapse
|
10
|
Wang Y, Wang Y, Yue G, Zhao Y. Energy metabolism disturbance in migraine: From a mitochondrial point of view. Front Physiol 2023; 14:1133528. [PMID: 37123270 PMCID: PMC10133718 DOI: 10.3389/fphys.2023.1133528] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Migraine is a serious central nervous system disease with a high incidence rate. Its pathogenesis is very complex, which brings great difficulties for clinical treatment. Recently, many studies have revealed that mitochondrial dysfunction may play a key role in migraine, which affects the hyperosmotic of Ca2+, the excessive production of free radicals, the decrease of mitochondrial membrane potential, the imbalance of mPTP opening and closing, and the decrease of oxidative phosphorylation level, which leads to neuronal energy exhaustion and apoptosis, and finally lessens the pain threshold and migraine attack. This article mainly introduces cortical spreading depression, a pathogenesis of migraine, and then damages the related function of mitochondria, which leads to migraine. Oxidative phosphorylation and the tricarboxylic acid cycle are the main ways to provide energy for the body. 95 percent of the energy needed for cell survival is provided by the mitochondrial respiratory chain. At the same time, hypoxia can lead to cell death and migraine. The pathological opening of the mitochondrial permeability transition pore can promote the interaction between pro-apoptotic protein and mitochondrial, destroy the structure of mPTP, and further lead to cell death. The increase of mPTP permeability can promote the accumulation of reactive oxygen species, which leads to a series of changes in the expression of proteins related to energy metabolism. Both Nitric oxide and Calcitonin gene-related peptide are closely related to the attack of migraine. Recent studies have shown that changes in their contents can also affect the energy metabolism of the body, so this paper reviews the above mechanisms and discusses the mechanism of brain energy metabolism of migraine, to provide new strategies for the prevention and treatment of migraine and promote the development of individualized and accurate treatment of migraine.
Collapse
Affiliation(s)
- Yicheng Wang
- Department of Neurology, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yongli Wang
- Department of Neurology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, China
| | - Guangxin Yue
- Institute of Basic Theory for Chinese Medicine, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yonglie Zhao
- Department of Neurology, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yonglie Zhao,
| |
Collapse
|
11
|
Park KY, Hong S, Kim KS, Han K, Park CY. Prolonged Use of Carnitine-Orotate Complex (Godex ®) Is Associated with Improved Mortality: A Nationwide Cohort Study. J Pers Med 2022; 12:jpm12121970. [PMID: 36556191 PMCID: PMC9787718 DOI: 10.3390/jpm12121970] [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: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Despite its hepatoprotective effects and favorable metabolic effects, the association between carnitine-orotate complex (Godex®) intake and mortality has never been investigated. We enrolled 13,413 adults who underwent national health examination and were prescribed the carnitine-orotate complex. Subjects were classified into three groups based on duration of using carnitine-orotate complex: <30, 30−180, and ≥180 days and were followed-up until 2019. Hazard ratios (HRs) and 95% confidence intervals (CIs) for all-cause mortality were estimated using Cox proportional hazards regression. During the follow-up period, 708 deaths were documented. Adjusted HR of mortality was 0.69 (95% CI 0.51−0.92) in those who used carnitine-orotate complex for ≥180 days compared to those who used it for <30 days. Use of carnitine-orotate complex for ≥180 days was associated with significantly reduced mortality in individuals with metabolic risk factors such as obesity, metabolic syndrome, dyslipidemia, and fatty liver than the shorter period of use. A significant interaction was observed in individuals with type 2 diabetes (HR 0.43, 95% CI 0.29−0.63, p-value 0.001). In this nationwide study, longer use of carnitine-orotate complex was associated with improved mortality compared to a shorter period of use, and the risk reductions were prominent in individuals with metabolic risk factors.
Collapse
Affiliation(s)
- Kye-Yeung Park
- Department of Family Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Sangmo Hong
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 11923, Republic of Korea
| | - Kyung-Soo Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13497, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Cheol-Young Park
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea
- Correspondence: ; Tel.: +82-2-2001-1869; Fax: +82-2001-1588
| |
Collapse
|
12
|
Bhattacharya S, Horowitz BB, Zhang J, Li X, Zhang H, Giese GE, Holdorf AD, Walhout AJ. A metabolic regulatory network for the Caenorhabditis elegans intestine. iScience 2022; 25:104688. [PMID: 35847555 PMCID: PMC9283940 DOI: 10.1016/j.isci.2022.104688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/12/2022] [Accepted: 06/24/2022] [Indexed: 11/12/2022] Open
Abstract
Metabolic perturbations can affect gene expression, for instance to rewire metabolism. While numerous efforts have measured gene expression in response to individual metabolic perturbations, methods that determine all metabolic perturbations that affect the expression for a given gene or set of genes have not been available. Here, we use a gene-centered approach to derive a first-pass metabolic regulatory network for Caenorhabditis elegans by performing RNAi of more than 1,400 metabolic genes with a set of 19 promoter reporter strains that express a fluorescent protein in the animal's intestine. We find that metabolic perturbations generally increase promoter activity, which contrasts with transcription factor (TF) RNAi, which tends to repress promoter activity. We identify several TFs that modulate promoter activity in response to perturbations of the electron transport chain and explore complex genetic interactions among metabolic pathways. This work provides a blueprint for a systems-level understanding of how metabolism affects gene expression.
Collapse
Affiliation(s)
- Sushila Bhattacharya
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Brent B. Horowitz
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jingyan Zhang
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xuhang Li
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hefei Zhang
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Gabrielle E. Giese
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Amy D. Holdorf
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Albertha J.M. Walhout
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
13
|
El Gaafary M, Saber FR, Mahrous EA, Ashour RM, Okba MM, Jin L, Lang SJ, Schmiech M, Simmet T, Syrovets T. The phloroglucinol calcitrinone A, a novel mitochondria-targeting agent, induces cell death in breast cancer cells. Food Chem Toxicol 2022; 162:112896. [PMID: 35227860 DOI: 10.1016/j.fct.2022.112896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/06/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
Abstract
Breast cancer is the most common cancer and the leading cause of cancer-related mortality among females worldwide. From the leaves of Callistemon citrinus, we have isolated a novel phloroglucinol dimer, calcitrinone A, and analyzed its potential anticancer activity using the triple-negative breast cancer cell line MDA-MB-231. Calcitrinone A decreased the total intracellular ATP levels, inhibited proliferation, and induced apoptosis in MDA-MB-231 cells, but was less toxic to peripheral blood mononuclear cells. The antiproliferative and apoptosis-inducing effects of calcitrinone A were confirmed in vivo using breast cancer xenografts grown on chick chorioallantoic membranes. Mechanistic analysis showed mitochondrial membrane-potential dissipation and interference with energy-yielding processes resulting in cell accumulation in the S phase of the cell cycle. Seahorse assay analysis revealed an early inhibition of mitochondrial oxidative phosphorylation (OXPHOS). At the molecular level, calcitrinone A inhibited activity of the succinate-coenzyme Q reductase (SQR) (mitochondrial complex II). In silico docking identified the coenzyme Q binding pocket as a possible high affinity binding site for calcitrinone A in SQR. Inhibition of complex II was accompanied by strong elevation of mitochondrial superoxide and cytoplasmic ROS. Calcitrinone A might be a promising anticancer lead compound acting through the interference with the mitochondrial complex II activity.
Collapse
Affiliation(s)
- Menna El Gaafary
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, 11562, Egypt; Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, 89081, Ulm, Germany
| | - Fatema R Saber
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Engy A Mahrous
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Rehab M Ashour
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mona M Okba
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Lu Jin
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, 89081, Ulm, Germany
| | - Sophia J Lang
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, 89081, Ulm, Germany
| | - Michael Schmiech
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, 89081, Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, 89081, Ulm, Germany.
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
14
|
Fujiwara H, Seike K, Brooks MD, Mathew AV, Kovalenko I, Pal A, Lee HJ, Peltier D, Kim S, Liu C, Oravecz-Wilson K, Li L, Sun Y, Byun J, Maeda Y, Wicha MS, Saunders TL, Rehemtulla A, Lyssiotis CA, Pennathur S, Reddy P. Mitochondrial complex II in intestinal epithelial cells regulates T cell-mediated immunopathology. Nat Immunol 2021; 22:1440-1451. [PMID: 34686860 PMCID: PMC9351914 DOI: 10.1038/s41590-021-01048-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/14/2021] [Indexed: 01/20/2023]
Abstract
Intestinal epithelial cell (IEC) damage by T cells contributes to graft-versus-host disease, inflammatory bowel disease and immune checkpoint blockade-mediated colitis. But little is known about the target cell-intrinsic features that affect disease severity. Here we identified disruption of oxidative phosphorylation and an increase in succinate levels in the IECs from several distinct in vivo models of T cell-mediated colitis. Metabolic flux studies, complemented by imaging and protein analyses, identified disruption of IEC-intrinsic succinate dehydrogenase A (SDHA), a component of mitochondrial complex II, in causing these metabolic alterations. The relevance of IEC-intrinsic SDHA in mediating disease severity was confirmed by complementary chemical and genetic experimental approaches and validated in human clinical samples. These data identify a critical role for the alteration of the IEC-specific mitochondrial complex II component SDHA in the regulation of the severity of T cell-mediated intestinal diseases.
Collapse
Affiliation(s)
- Hideaki Fujiwara
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Keisuke Seike
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Michael D Brooks
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Anna V Mathew
- Department of Internal Medicine, Division of Nephrology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Ilya Kovalenko
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Anupama Pal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ho-Joon Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Peltier
- Department of Pediatrics, Division of Hematology/Oncology and BMT, University of Michigan Health System, Ann Arbor, MI, USA
| | - Stephanie Kim
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Katherine Oravecz-Wilson
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Lu Li
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yaping Sun
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jaeman Byun
- Department of Internal Medicine, Division of Nephrology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Yoshinobu Maeda
- Department of Hematology Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Max S Wicha
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine, Division of Nephrology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Pavan Reddy
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Moerman F, Fronhofer EA, Altermatt F, Wagner A. Selection on growth rate and local adaptation drive genomic adaptation during experimental range expansions in the protist Tetrahymena thermophila. J Anim Ecol 2021; 91:1088-1103. [PMID: 34582573 PMCID: PMC9291582 DOI: 10.1111/1365-2656.13598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/19/2021] [Indexed: 11/29/2022]
Abstract
Populations that expand their range can undergo rapid evolutionary adaptation of life‐history traits, dispersal behaviour and adaptation to the local environment. Such adaptation may be aided or hindered by sexual reproduction, depending on the context. However, few empirical and experimental studies have investigated the genetic basis of adaptive evolution during range expansions. Even less attention has been given to the question how sexual reproduction may modulate such adaptive evolution during range expansions. We here studied genomic adaptation during experimental range expansions of the protist Tetrahymena thermophila in landscapes with a uniform environment or a pH gradient. Specifically, we investigated two aspects of genomic adaptation during range expansion. First, we investigated adaptive genetic change in terms of the underlying numbers of allele frequency changes from standing genetic variation and de novo variants. We focused on how sexual reproduction may alter this adaptive genetic change. Second, we identified genes subject to selection caused by the expanding range itself, and directional selection due to the presence or absence of the pH gradient. We focused this analysis on alleles with large frequency changes that occurred in parallel in more than one population to identify the most likely candidate targets of selection. We found that sexual reproduction altered adaptive genetic change both in terms of de novo variants and standing genetic variation. However, sexual reproduction affected allele frequency changes in standing genetic variation only in the absence of long‐distance gene flow. Adaptation to the range expansion affected genes involved in cell divisions and DNA repair, whereas adaptation to the pH gradient additionally affected genes involved in ion balance and oxidoreductase reactions. These genetic changes may result from selection on growth and adaptation to low pH. In the absence of gene flow, sexual reproduction may have aided genetic adaptation. Gene flow may have swamped expanding populations with maladapted alleles, thus reducing the extent of evolutionary adaptation during range expansion. Sexual reproduction also altered the genetic basis of adaptation in our evolving populations via de novo variants, possibly by purging deleterious mutations or by revealing fitness benefits of rare genetic variants.
Collapse
Affiliation(s)
- Felix Moerman
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland.,Department of Aquatic Ecology, Eawag: Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland.,Swiss Institute of Bioinformatics, Quartier Sorge-Bâtiment Génopode, Lausanne, Switzerland.,ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | | | - Florian Altermatt
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland.,Department of Aquatic Ecology, Eawag: Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Andreas Wagner
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland.,Swiss Institute of Bioinformatics, Quartier Sorge-Bâtiment Génopode, Lausanne, Switzerland.,The Santa Fe Institute, Santa Fe, NM, USA.,Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
16
|
Fedotcheva TA, Fedotcheva NI. Protectors of the Mitochondrial Permeability Transition Pore Activated by Iron and Doxorubicin. Curr Cancer Drug Targets 2021; 21:514-525. [PMID: 33475063 DOI: 10.2174/1568009621999210120192558] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/22/2022]
Abstract
AIM The study is aimed at examining of action of iron, DOX, and their complex on the Mitochondrial Permeability Transition Pore (MPTP) opening and detecting of possible protectors of MPTP in the conditions close to mitochondria-dependent ferroptosis. BACKGROUND The Toxicity of Doxorubicin (DOX) is mainly associated with free iron accumulation and mitochondrial dysfunction. DOX can provoke ferroptosis, iron-dependent cell death driven by membrane damage. The Mitochondrial Permeability Transition Pore (MPTP) is considered as a common pathway leading to the development of apoptosis, necrosis, and, possibly, ferroptosis. The influence of DOX on the Ca2+ -induced MPTP opening in the presence of iron has not yet been studied. OBJECTIVE The study was conducted on isolated liver and heart mitochondria. MPTP and succinate- ubiquinone oxidoreductase were studied as targets of DOX in mitochondria-dependent ferroptosis. The iron chelator deferoxamine (DFO), the lipid radical scavenger butyl-hydroxytoluene (BHT), and rutenium red (Rr), as a possible inhibitor of ferrous ions uptake in mitochondria, were tested as MPTP protectors. The role of medium alkalization was also examined. METHODS Changes of threshold calcium concentrations required for MPTP opening were measured by a Ca2+ selective electrode, mitochondrial membrane potential was registered by tetraphenylphosphonium (TPP+)-selective electrode, and mitochondrial swelling was recorded as a decrease in absorbance at 540 nm. The activity of Succinate Dehydrogenase (SDH) was determined by the reduction of the electron acceptor DCPIP. CONCLUSION MPTP and the respiratory complex II are identified as the main targets of the iron-dependent action of DOX on the isolated mitochondria. All MPTP protectors tested abolished or weakened the effect of iron and a complex of iron with DOX on Ca2+ -induced MPTP opening, acting in different stages of MPTP activation. These data open new approaches to the modulation of the toxic influence of DOX on mitochondria with the aim to reduce their dysfunction.
Collapse
Affiliation(s)
- Tatiana A Fedotcheva
- Science Research Laboratory of Pharmacology, Faculty of Medical Biology, N. I. Pirogov Russian National Medical Research University, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Nadezhda I Fedotcheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, 142290, Russian Federation
| |
Collapse
|
17
|
Hadrava Vanova K, Kraus M, Neuzil J, Rohlena J. Mitochondrial complex II and reactive oxygen species in disease and therapy. Redox Rep 2021; 25:26-32. [PMID: 32290794 PMCID: PMC7178880 DOI: 10.1080/13510002.2020.1752002] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Increasing evidence points to the respiratory Complex II (CII) as a source and modulator of reactive oxygen species (ROS). Both functional loss of CII as well as its pharmacological inhibition can lead to ROS generation in cells, with a relevant impact on the development of pathophysiological conditions, i.e. cancer and neurodegenerative diseases. While the basic framework of CII involvement in ROS production has been defined, the fine details still await clarification. It is important to resolve these aspects to fully understand the role of CII in pathology and to explore its therapeutic potential in cancer and other diseases.
Collapse
Affiliation(s)
| | - Michal Kraus
- Institute of Biotechnology of the Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology of the Czech Academy of Sciences, Prague-West, Czech Republic.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - Jakub Rohlena
- Institute of Biotechnology of the Czech Academy of Sciences, Prague-West, Czech Republic
| |
Collapse
|
18
|
Saxena N, Beraldi E, Fazli L, Somasekharan SP, Adomat H, Zhang F, Molokwu C, Gleave A, Nappi L, Nguyen K, Brar P, Nikesitch N, Wang Y, Collins C, Sorensen PH, Gleave M. Androgen receptor (AR) antagonism triggers acute succinate-mediated adaptive responses to reactivate AR signaling. EMBO Mol Med 2021; 13:e13427. [PMID: 33709547 PMCID: PMC8103094 DOI: 10.15252/emmm.202013427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 11/09/2022] Open
Abstract
Treatment-induced adaptive pathways converge to support androgen receptor (AR) reactivation and emergence of castration-resistant prostate cancer (PCa) after AR pathway inhibition (ARPI). We set out to explore poorly defined acute adaptive responses that orchestrate shifts in energy metabolism after ARPI and identified rapid changes in succinate dehydrogenase (SDH), a TCA cycle enzyme with well-known tumor suppressor activity. We show that AR directly regulates transcription of its catalytic subunits (SDHA, SDHB) via androgen response elements (AREs). ARPI acutely suppresses SDH activity, leading to accumulation of the oncometabolite, succinate. Succinate triggers calcium ions release from intracellular stores, which in turn phospho-activates the AR-cochaperone, Hsp27 via p-CaMKK2/p-AMPK/p-p38 axis to enhance AR protein stabilization and activity. Activation of this pathway was seen in tissue microarray analysis on prostatectomy tissues and patient-derived xenografts. This adaptive response is blocked by co-targeting AR with Hsp27 under both in vitro and in vivo studies, sensitizing PCa cells to ARPI treatments.
Collapse
Affiliation(s)
- Neetu Saxena
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Hans Adomat
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Fan Zhang
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Anna Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lucia Nappi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Pavn Brar
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Adiponectin enhances the bioenergetics of cardiac myocytes via an AMPK- and succinate dehydrogenase-dependent mechanism. Cell Signal 2021; 78:109866. [PMID: 33271223 PMCID: PMC9619024 DOI: 10.1016/j.cellsig.2020.109866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Adiponectin is one of the most abundant circulating hormones, which through adenosine monophosphate-activated protein kinase (AMPK), enhances fatty acid and glucose oxidation, and exerts a cardioprotective effect. However, its effects on cellular bioenergetics have not been explored. We have previously reported that 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR, an AMPK activator) enhances mitochondrial respiration through a succinate dehydrogenase (SDH or complex II)-dependent mechanism in cardiac myocytes, leading us to predict that Adiponectin would exert a similar effect via activating AMPK. Our results show that Adiponectin enhances basal mitochondrial oxygen consumption rate (OCR), ATP production, and spare respiratory capacity (SRC), which were all abolished by the knockdown of AMPKγ1, inhibition of SDH complex assembly, via the knockdown of the SDH assembly factor 1 (Sdhaf1), or inhibition of SDH activity. Additionally, Adiponectin alleviated hypoxia-induced reductions in OCR and ATP production, in a Sdhaf1-dependent manner, whereas overexpression of Sdhaf1 confirmed its sufficiency for mediating these effects. Importantly, the levels of holoenzyme SDH under the various conditions correlated with OCR. We also show that the effects of Adiponectin, AMPK, Sdhaf1, as well as, SDH complex assembly all required sirtuin 3 (Sirt3). In conclusion, Adiponectin potentiates mitochondrial bioenergetics via promoting SDH complex assembly in an AMPK-, Sdhaf1-, and Sirt3-dependent fashion in cardiac myocytes.
Collapse
|
20
|
Markevich NI, Markevich LN, Hoek JB. Computational Modeling Analysis of Generation of Reactive Oxygen Species by Mitochondrial Assembled and Disintegrated Complex II. Front Physiol 2020; 11:557721. [PMID: 33178032 PMCID: PMC7596731 DOI: 10.3389/fphys.2020.557721] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/31/2020] [Indexed: 11/13/2022] Open
Abstract
Reactive oxygen species (ROS) function as critical mediators in a broad range of cellular signaling processes. The mitochondrial electron transport chain is one of the major contributors to ROS formation in most cells. Increasing evidence indicates that the respiratory Complex II (CII) can be the predominant ROS generator under certain conditions. A computational, mechanistic model of electron transfer and ROS formation in CII was developed in the present study to facilitate quantitative analysis of mitochondrial ROS production. The model was calibrated by fitting the computer simulated results to experimental data obtained on submitochondrial particles (SMP) prepared from bovine and rat heart mitochondria upon inhibition of the ubiquinone (Q)-binding site by atpenin A5 (AA5) and Complex III by myxothiazol, respectively. The model predicts that only reduced flavin adenine dinucleotide (FADH2) in the unoccupied dicarboxylate state and flavin semiquinone radical (FADH•) feature the experimentally observed bell-shaped dependence of the rate of ROS production on the succinate concentration upon inhibition of respiratory Complex III (CIII) or Q-binding site of CII, i.e., suppression of succinate-Q reductase (SQR) activity. The other redox centers of CII such as Fe-S clusters and Q-binding site have a hyperbolic dependence of ROS formation on the succinate concentration with very small maximal rate under any condition and cannot be considered as substantial ROS generators in CII. Computer simulation results show that CII disintegration (which results in dissociation of the hydrophilic SDHA/SDHB subunits from the inner membrane to the mitochondrial matrix) causes crucial changes in the kinetics of ROS production by CII that are qualitatively and quantitatively close to changes in the kinetics of ROS production by assembled CII upon inhibition of CIII or Q-binding site of CII. Thus, the main conclusions from the present computational modeling study are the following: (i) the impairment of the SQR activity of CII resulting from inhibition of CIII or Q-binding site of CII and (ii) CII disintegration causes a transition in the succinate-dependence of ROS production from a small-amplitude sigmoid (hyperbolic) shape, determined by Q-binding site or [3Fe-4S] cluster to a high-amplitude bell-shaped kinetics with a shift to small subsaturated concentrations of succinate, determined by the flavin site.
Collapse
Affiliation(s)
| | | | - Jan B Hoek
- MitoCare Center for Mitochondrial Research, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
21
|
Huang Z, Chen Y, Zhang Y. Mitochondrial reactive oxygen species cause major oxidative mitochondrial DNA damages and repair pathways. J Biosci 2020. [DOI: 10.1007/s12038-020-00055-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Laustsen C, Nielsen PM, Qi H, Løbner MH, Palmfeldt J, Bertelsen LB. Hyperpolarized [1,4- 13C]fumarate imaging detects microvascular complications and hypoxia mediated cell death in diabetic nephropathy. Sci Rep 2020; 10:9650. [PMID: 32541797 PMCID: PMC7295762 DOI: 10.1038/s41598-020-66265-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Today, there is a general lack of prognostic biomarkers for development of renal disease and in particular diabetic nephropathy. Increased glycolytic activity, lactate accumulation and altered mitochondrial oxygen utilization are hallmarks of diabetic kidney disease. Fumarate hydratase activity has been linked to mitochondrial dysfunction as well as activation of the hypoxia inducible factor, induction of apoptosis and necrosis. Here, we investigate fumarate hydratase activity in biofluids in combination with the molecular imaging probe, hyperpolarized [1,4-13C2]fumarate, to identify the early changes associated with hemodynamics and cell death in a streptozotocin rat model of type 1 diabetes. We found a significantly altered hemodynamic signature of [1,4-13C2]fumarate in the diabetic kidneys as well as an systemic increased metabolic conversion of fumarate-to-malate, indicative of increased cell death associated with progression of diabetes, while little to no renal specific conversion was observed. This suggest apoptosis as the main cause of cell death in the diabetic kidney. This is likely resulting from an increased reactive oxygen species production following uncoupling of the electron transport chain at complex II. The mechanism coupling the enzyme leakage and apoptotic phenotype is hypoxia inducible factor independent and seemingly functions as a protective mechanism in the kidney cells.
Collapse
Affiliation(s)
- Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Per Mose Nielsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Haiyun Qi
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mette Hadberg Løbner
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lotte Bonde Bertelsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
23
|
Heng T, Kaga A, Chen X, Somta P. Two tightly linked genes coding for NAD-dependent malic enzyme and dynamin-related protein are associated with resistance to Cercospora leaf spot disease in cowpea (Vigna unguiculata (L.) Walp.). TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2020; 133:395-407. [PMID: 31691838 DOI: 10.1007/s00122-019-03470-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 10/28/2019] [Indexed: 05/20/2023]
Abstract
Cercospora leaf spot (CLS) caused by Cercospora canescens is an important disease of cowpea (Vigna unguiculata). A previous study using an F2 population [CSR12906 (susceptible) × IT90K-59-120 (resistant)] identified a major QTL qCLS9.1 for resistance to CLS. In this study, we finely mapped and identified candidate genes of qCLS9.1 using an F3:4 population of 699 individuals derived from two F2:3 individuals segregating at qCLS9.1 from the original population. Fine mapping narrowed down the qCLS9.1 for the resistance to a 60.6-Kb region on cowpea chromosome 10. There were two annotated genes in the 60.6-Kb region; Vigun10g019300 coding for NAD-dependent malic enzyme 1 (NAD-ME1) and Vigun10g019400 coding for dynamin-related protein 1C (DRP1C). DNA sequence analysis revealed 12 and 2 single nucleotide polymorphisms (SNPs) in the coding sequence (CDS) and the 5' untranslated region and TATA boxes of Vigun10g019300 and Vigun10g019400, respectively. Three SNPs caused amino acid changes in NAD-ME1 in CSR12906, N299S, S488N and S544N. Protein prediction analysis suggested that S488N of CSR12906 may have a deleterious effect on the function of NAD-ME1. Gene expression analysis demonstrated that IT90K-59-120 and CSR12906 challenged with C. canescens showed different expression in both Vigun10g019300 and Vigun10g019400. Taken together, these results indicated that Vigun10g019300 and Vigun10g019400 are the candidate genes for CLS resistance in the cowpea IT90K-59-120. Two derived cleaved amplified polymorphic sequence markers were developed to detect the resistance alleles at Vigun10g019300 and Vigun10g019400 in IT90K-59-120.
Collapse
Affiliation(s)
- Titnarong Heng
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Kamphaeng Saen, Nakhon Pathom, 73140, Thailand
| | - Akito Kaga
- Soybean and Field Crop Applied Genomics Research Unit, Institute of Crop Science, National Agriculture and Food Research Organization, 2-1-2, Kannondai, Tsukuba, Ibaraki, 305-8602, Japan
| | - Xin Chen
- Institute of Industrial Crops, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, Jiangsu, China
| | - Prakit Somta
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Kamphaeng Saen, Nakhon Pathom, 73140, Thailand.
- Center of Excellence on Agricultural Biotechnology: (AG-BIO/PERDO-CHE), Bangkok, 10900, Thailand.
| |
Collapse
|
24
|
Okoye CN, MacDonald-Jay N, Kamunde C. Effects of bioenergetics, temperature and cadmium on liver mitochondria reactive oxygen species production and consumption. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 214:105264. [PMID: 31377504 DOI: 10.1016/j.aquatox.2019.105264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 06/10/2023]
Abstract
A by-product of mitochondrial substrate oxidation and electron transfer to generate cellular energy (ATP) is reactive oxygen species (ROS). Superoxide anion radical and hydrogen peroxide (H2O2) are the proximal ROS produced by the mitochondria. Because low levels of ROS serve critical regulatory roles in cell physiology while excessive levels or inappropriately localized ROS result in aberrant physiological states, mitochondrial ROS need to be tightly regulated. While it is known that regulation of mitochondrial ROS involves balancing the rates of production and removal, the effects of stressors on these processes remain largely unknown. To illuminate how stressors modulate mitochondrial ROS homeostasis, we investigated the effects of temperature and cadmium (Cd) on H2O2 emission and consumption in rainbow trout liver mitochondria. We show that H2O2 emission rates increase with temperature and Cd exposure. Energizing mitochondria with malate-glutamate or succinate increased the rate of H2O2 emission; however, Cd exposure imposed different patterns of H2O2 emission depending on the concentration and substrate. Specifically, mitochondria respiring on malate-glutamate exhibited a saturable graded concentration-response curve that plateaued at 5 μM while mitochondria respiring on succinate had a biphasic concentration-response curve characterized by a spike in the emission rate at 1 μM Cd followed by gradual diminution at higher Cd concentrations. To explain the observed substrate- and concentration-dependent effects of Cd, we sequestered specific mitochondrial ROS-emitting sites using blockers of electron transfer and then tested the effect of the metal. The results indicate that the biphasic H2O2 emission response imposed by succinate is due to site IIF but is further modified at sites IQ and IIIQo. Moreover, the saturable graded H2O2 emission response in mitochondria energized with malate-glutamate is consistent with effect of Cd on site IF. Additionally, Cd and temperature acted cooperatively to increase mitochondrial H2O2 emission suggesting that increased toxicity of Cd at high temperature may be due to increased oxidative insult. Surprisingly, despite their clear stimulatory effect on H2O2 emission, Cd, temperature and bioenergetic status did not affect the kinetics of mitochondrial H2O2 consumption; the rate constants and half-lives for all the conditions tested were similar. Overall, our study indicates that the production processes of rainbow trout liver mitochondrial H2O2 metabolism are highly responsive to stressors and bioenergetics while the consumption processes are recalcitrant. The latter denotes the presence of a robust H2O2 scavenging system in liver mitochondria that would maintain H2O2 homeostasis in the face of increased production and reduced scavenging capacity.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| | - Nicole MacDonald-Jay
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada.
| |
Collapse
|
25
|
Lagadic-Gossmann D, Hardonnière K, Mograbi B, Sergent O, Huc L. Disturbances in H + dynamics during environmental carcinogenesis. Biochimie 2019; 163:171-183. [PMID: 31228544 DOI: 10.1016/j.biochi.2019.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/16/2019] [Indexed: 12/24/2022]
Abstract
Despite the improvement of diagnostic methods and anticancer therapeutics, the human population is still facing an increasing incidence of several types of cancers. According to the World Health Organization, this growing trend would be partly linked to our environment, with around 20% of cancers stemming from exposure to environmental contaminants, notably chemicals like polycyclic aromatic hydrocarbons (PAHs). PAHs are widespread pollutants in our environment resulting from incomplete combustion or pyrolysis of organic material, and thus produced by both natural and anthropic sources; notably benzo[a]pyrene (B[a]P), i.e. the prototypical molecule of this family, that can be detected in cigarette smoke, diesel exhaust particles, occupational-related fumes, and grilled food. This molecule is a well-recognized carcinogen belonging to group 1 carcinogens. Indeed, it can target the different steps of the carcinogenic process and all cancer hallmarks. Interestingly, H+ dynamics have been described as key parameters for the occurrence of several, if not all, of these hallmarks. However, information regarding the role of such parameters during environmental carcinogenesis is still very scarce. The present review will thus mainly give an overview of the impact of B[a]P on H+ dynamics in liver cells, and will show how such alterations might impact different aspects related to the finely-tuned balance between cell death and survival processes, thereby likely favoring environmental carcinogenesis. In total, the main objective of this review is to encourage further research in this poorly explored field of environmental molecular toxicology.
Collapse
Affiliation(s)
- Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France.
| | - Kévin Hardonnière
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Baharia Mograbi
- Institute of Research on Cancer and Ageing of Nice (IRCAN), INSERM U1081, CNRS UMR7284, 2. Université de Nice-Sophia Antipolis, Faculté de Médecine, Centre Antoine Lacassagne, Nice, F-06107, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Laurence Huc
- INRA, ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
26
|
Ralph SJ, Nozuhur S, ALHulais RA, Rodríguez‐Enríquez S, Moreno‐Sánchez R. Repurposing drugs as pro‐oxidant redox modifiers to eliminate cancer stem cells and improve the treatment of advanced stage cancers. Med Res Rev 2019; 39:2397-2426. [DOI: 10.1002/med.21589] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/20/2019] [Accepted: 03/31/2019] [Indexed: 01/10/2023]
Affiliation(s)
- Stephen J. Ralph
- School of Medical ScienceGriffith University Southport Australia
| | - Sam Nozuhur
- School of Medical ScienceGriffith University Southport Australia
| | | | | | | |
Collapse
|
27
|
Investigating the Effects of Stove Emissions on Ocular and Cancer Cells. Sci Rep 2019; 9:1870. [PMID: 30755694 PMCID: PMC6372759 DOI: 10.1038/s41598-019-38803-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022] Open
Abstract
More than a third of the world’s population relies on solid fuels for cooking and heating, with major health consequences. Although solid fuel combustion emissions are known to increase the prevalence of illnesses such as chronic obstructive pulmonary disease and lung cancer, however, their effect on the eyes is underexplored. This study assesses the acute toxicity of solid fuel combustion emissions on healthy ocular cells and a cancer cell line. Three healthy ocular cell lines (corneal, lens, and retinal epithelial cells) and a cancer cell line (Chinese hamster ovary cells) were exposed to liquid and gas phase emissions from applewood and coal combustion. Following the exposure, real-time cell attachment behavior was monitored for at least 120 hours with electrical cell impedance spectroscopy. The viability of the cells, amount of apoptotic cells, and generation of reactive oxygen species (ROS) were quantified with MTT, ApoTox-Glo, and ROS-Glo H2O2 assays, respectively. The results showed that coal emissions compromised the viability of ocular cells more than applewood emissions. Interestingly, the cancer cells, although their viability was not compromised, generated 1.7 to 2.7 times more ROS than healthy cells. This acute exposure study provides compelling proof that biomass combustion emissions compromise the viability of ocular cells and increase ROS generation. The increased ROS generation was fatal for ocular cells, but it promoted the growth of cancer cells.
Collapse
|
28
|
Devaux JBL, Hedges CP, Birch N, Herbert N, Renshaw GMC, Hickey AJR. Acidosis Maintains the Function of Brain Mitochondria in Hypoxia-Tolerant Triplefin Fish: A Strategy to Survive Acute Hypoxic Exposure? Front Physiol 2019; 9:1941. [PMID: 30713504 PMCID: PMC6346031 DOI: 10.3389/fphys.2018.01941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/22/2018] [Indexed: 11/13/2022] Open
Abstract
The vertebrate brain is generally very sensitive to acidosis, so a hypoxia-induced decrease in pH is likely to have an effect on brain mitochondria (mt). Mitochondrial respiration (JO2) is required to generate an electrical gradient (ΔΨm) and a pH gradient to power ATP synthesis, yet the impact of pH modulation on brain mt function remains largely unexplored. As intertidal fishes within rock pools routinely experience hypoxia and reoxygenation, they would most likely experience changes in cellular pH. We hence compared four New Zealand triplefin fish species ranging from intertidal hypoxia-tolerant species (HTS) to subtidal hypoxia-sensitive species (HSS). We predicted that HTS would tolerate acidosis better than HSS in terms of sustaining mt structure and function. Using respirometers coupled to fluorimeters and pH electrodes, we titrated lactic-acid to decrease the pH of the media, and simultaneously recorded JO2, ΔΨm, and H+ buffering capacities within permeabilized brain and swelling of mt isolated from non-permeabilized brains. We then measured ATP synthesis rates in the most HTS (Bellapiscus medius) and the HSS (Forsterygion varium) at pH 7.25 and 6.65. Mitochondria from HTS brain did have greater H+ buffering capacities than HSS mt (∼10 mU pH.mgprotein -1). HTS mt swelled by 40% when exposed to a decrease of 1.5 pH units, and JO2 was depressed by up to 15% in HTS. However, HTS were able to maintain ΔΨm near -120 mV. Estimates of work, in terms of charges moved across the mt inner-membrane, suggested that with acidosis, HTS mt may in part harness extra-mt H+ to maintain ΔΨm, and could therefore support ATP production. This was confirmed with elevated ATP synthesis rates and enhanced P:O ratios at pH 6.65 relative to pH 7.25. In contrast, mt volumes and ΔΨm decreased downward pH 6.9 in HSS mt and paradoxically, JO2 increased (∼25%) but ATP synthesis and P:O ratios were depressed at pH 6.65. This indicates a loss of coupling in the HSS with acidosis. Overall, the mt of these intertidal fish have adaptations that enhance ATP synthesis efficiency under acidic conditions such as those that occur in hypoxic or reoxygenated brain.
Collapse
Affiliation(s)
- Jules B L Devaux
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Christopher P Hedges
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Nigel Birch
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Neill Herbert
- Institute of Marine Science, The University Auckland, Auckland, New Zealand
| | - Gillian M C Renshaw
- School of Allied Health Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Anthony J R Hickey
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
Russo E, Nguyen H, Lippert T, Tuazon J, Borlongan CV, Napoli E. Mitochondrial targeting as a novel therapy for stroke. Brain Circ 2018; 4:84-94. [PMID: 30450413 PMCID: PMC6187947 DOI: 10.4103/bc.bc_14_18] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 01/16/2023] Open
Abstract
Stroke is a main cause of mortality and morbidity worldwide. Despite the increasing development of innovative treatments for stroke, most are unsuccessful in clinical trials. In recent years, an encouraging strategy for stroke therapy has been identified in stem cells transplantation. In particular, grafting cells and their secretion products are leading with functional recovery in stroke patients by promoting the growth and function of the neurovascular unit – a communication framework between neurons, their supply microvessels along with glial cells – underlying stroke pathology and recovery. Mitochondrial dysfunction has been recently recognized as a hallmark in ischemia/reperfusion neural damage. Emerging evidence of mitochondria transfer from stem cells to ischemic-injured cells points to transfer of healthy mitochondria as a viable novel therapeutic strategy for ischemic diseases. Hence, a more in-depth understanding of the cellular and molecular mechanisms involved in mitochondrial impairment may lead to new tools for stroke treatment. In this review, we focus on the current evidence of mitochondrial dysfunction in stroke, investigating favorable approaches of healthy mitochondria transfer in ischemic neurons, and exploring the potential of mitochondria-based cellular therapy for clinical applications. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed.
Collapse
Affiliation(s)
- Eleonora Russo
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Hung Nguyen
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Trenton Lippert
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Julian Tuazon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| |
Collapse
|
30
|
Understanding the Role of Dysfunctional and Healthy Mitochondria in Stroke Pathology and Its Treatment. Int J Mol Sci 2018; 19:ijms19072127. [PMID: 30037107 PMCID: PMC6073421 DOI: 10.3390/ijms19072127] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022] Open
Abstract
Stroke remains a major cause of death and disability in the United States and around the world. Solid safety and efficacy profiles of novel stroke therapeutics have been generated in the laboratory, but most failed in clinical trials. Investigations into the pathology and treatment of the disease remain a key research endeavor in advancing scientific understanding and clinical applications. In particular, cell-based regenerative medicine, specifically stem cell transplantation, may hold promise as a stroke therapy, because grafted cells and their components may recapitulate the growth and function of the neurovascular unit, which arguably represents the alpha and omega of stroke brain pathology and recovery. Recent evidence has implicated mitochondria, organelles with a central role in energy metabolism and stress response, in stroke progression. Recognizing that stem cells offer a source of healthy mitochondria—one that is potentially transferrable into ischemic cells—may provide a new therapeutic tool. To this end, deciphering cellular and molecular processes underlying dysfunctional mitochondria may reveal innovative strategies for stroke therapy. Here, we review recent studies capturing the intimate participation of mitochondrial impairment in stroke pathology, and showcase promising methods of healthy mitochondria transfer into ischemic cells to critically evaluate the potential of mitochondria-based stem cell therapy for stroke patients.
Collapse
|
31
|
Kume A, Kasai S, Furuya H, Suzuki H. α-Tocopheryl succinate-suppressed development of cerebral malaria in mice. Parasitol Res 2018; 117:3177-3182. [PMID: 30030625 DOI: 10.1007/s00436-018-6016-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022]
Abstract
α-Tocopheryl succinate (α-TOS), a derivative of vitamin E, is synthesized by esterification of α-tocopherol. It has been reported that α-TOS inhibits the mitochondrial complex II resulting in generation of reactive oxygen species, which triggers selective apoptosis in a large number of cancer cells, while it appears largely non-toxic towards normal cells. Plasmodium parasites are well known to have high sensitivity to oxidative stress. Thus, α-TOS is suspected to impact Plasmodium parasites by oxidative stress. In this study, to ascertain whether α-TOS is an appropriate candidate for an anti-malarial drug, C57BL/6J mice were infected with P. yoelii 17XL and P. berghei ANKA, a lethal strain of rodent malaria and experimental cerebral malaria (ECM), and treated with several concentrations of α-TOS by intraperitoneal administration on 1, 3, 5, and 7 days post infection (dpi). In addition, the permeability of the blood brain barrier (BBB) was examined by Evans blue staining in ECM on 7 dpi. As a result of α-TOS treatment, parasitemia was decreased and survival rate was significantly increased in mice infected with both parasites. Furthermore, the intensity of Evans blue staining on brains taken from α-TOS-treated mice was weaker than that of untreated mice. This means that α-TOS might inhibit the breakdown of BBB and progress of cerebral malaria. These findings indicate that vitamin E derivatives like α-TOS might be a potential candidate for treatment drugs against malaria.
Collapse
Affiliation(s)
- Aiko Kume
- Research Unit for Functional Genomics, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Shunji Kasai
- Eisai Co., Ltd., 13-1 Nishigoken-cho, Shinjuku-ku, Tokyo, 162-0812, Japan
| | - Hana Furuya
- Research Unit for Functional Genomics, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Hiroshi Suzuki
- Research Unit for Functional Genomics, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
32
|
Combination of Mitochondrial and Plasma Membrane Citrate Transporter Inhibitors Inhibits De Novo Lipogenesis Pathway and Triggers Apoptosis in Hepatocellular Carcinoma Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3683026. [PMID: 29546056 PMCID: PMC5818947 DOI: 10.1155/2018/3683026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/23/2017] [Accepted: 12/03/2017] [Indexed: 12/27/2022]
Abstract
Increased expression levels of both mitochondrial citrate transporter (CTP) and plasma membrane citrate transporter (PMCT) proteins have been found in various cancers. The transported citrates by these two transporter proteins provide acetyl-CoA precursors for the de novo lipogenesis (DNL) pathway to support a high rate of cancer cell viability and development. Inhibition of the DNL pathway promotes cancer cell apoptosis without apparent cytotoxic to normal cells, leading to the representation of selective and powerful targets for cancer therapy. The present study demonstrates that treatments with CTP inhibitor (CTPi), PMCT inhibitor (PMCTi), and the combination of CTPi and PMCTi resulted in decreased cell viability in two hepatocellular carcinoma cell lines (HepG2 and HuH-7). Treatment with citrate transporter inhibitors caused a greater cytotoxic effect in HepG2 cells than in HuH-7 cells. A lower concentration of combined CTPi and PMCTi promotes cytotoxic effect compared with either of a single compound. An increased cell apoptosis and an induced cell cycle arrest in both cell lines were reported after administration of the combined inhibitors. A combination treatment exhibits an enhanced apoptosis through decreased intracellular citrate levels, which consequently cause inhibition of fatty acid production in HepG2 cells. Apoptosis induction through the mitochondrial-dependent pathway was found as a consequence of suppressed carnitine palmitoyl transferase-1 (CPT-1) activity and enhanced ROS generation by combined CTPi and PMCTi treatment. We showed that accumulation of malonyl-CoA did not correlate with decreasing CPT-1 activity. The present study showed that elevated ROS levels served as an inhibition on Bcl-2 activity that is at least in part responsible for apoptosis. Moreover, inhibition of the citrate transporter is selectively cytotoxic to HepG2 cells but not in primary human hepatocytes, supporting citrate-mediating fatty acid synthesis as a promising cancer therapy.
Collapse
|
33
|
Idelchik MDPS, Begley U, Begley TJ, Melendez JA. Mitochondrial ROS control of cancer. Semin Cancer Biol 2017; 47:57-66. [PMID: 28445781 PMCID: PMC5653465 DOI: 10.1016/j.semcancer.2017.04.005] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 04/07/2017] [Accepted: 04/14/2017] [Indexed: 02/06/2023]
Abstract
Mitochondria serves a primary role in energy maintenance but also function to govern levels of mitochondria-derived reactive oxygen species (mROS). ROS have long been established to play a critical role in tumorigenesis and are now considered to be integral to the regulation of diverse signaling networks that drive proliferation, tumor cell survival and malignant progression. mROS can damage DNA, activate oncogenes, block the function of tumor suppressors and drive migratory signaling. The mitochondrion's oxidant scavenging systems including SOD2, Grx2, GPrx, Trx and TrxR are key of the cellular redox tone. These mitochondrial antioxidant systems serve to tightly control the levels of the primary ROS signaling species, H2O2. The coordinated control of mROS levels is also coupled to the activity of the primary H2O2 consuming enzymes of the mitochondria which are reliant on the epitranscriptomic control of selenocysteine incorporation. This review highlights the interplay between these many oncogenic signaling networks, mROS and the H2O2 emitting and consuming capacity of the mitochondria.
Collapse
Affiliation(s)
- María Del Pilar Sosa Idelchik
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States
| | - Ulrike Begley
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States
| | - Thomas J Begley
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States
| | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States.
| |
Collapse
|
34
|
Caino MC, Seo JH, Wang Y, Rivadeneira DB, Gabrilovich DI, Kim ET, Weeraratna AT, Languino LR, Altieri DC. Syntaphilin controls a mitochondrial rheostat for proliferation-motility decisions in cancer. J Clin Invest 2017; 127:3755-3769. [PMID: 28891816 DOI: 10.1172/jci93172] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/21/2017] [Indexed: 12/13/2022] Open
Abstract
Tumors adapt to an unfavorable microenvironment by controlling the balance between cell proliferation and cell motility, but the regulators of this process are largely unknown. Here, we show that an alternatively spliced isoform of syntaphilin (SNPH), a cytoskeletal regulator of mitochondrial movements in neurons, is directed to mitochondria of tumor cells. Mitochondrial SNPH buffers oxidative stress and maintains complex II-dependent bioenergetics, sustaining local tumor growth while restricting mitochondrial redistribution to the cortical cytoskeleton and tumor cell motility. Conversely, introduction of stress stimuli to the microenvironment, including hypoxia, acutely lowered SNPH levels, resulting in bioenergetics defects and increased superoxide production. In turn, this suppressed tumor cell proliferation but increased tumor cell invasion via greater mitochondrial trafficking to the cortical cytoskeleton. Loss of SNPH or expression of an SNPH mutant lacking the mitochondrial localization sequence resulted in increased metastatic dissemination in xenograft or syngeneic tumor models in vivo. Accordingly, tumor cells that acquired the ability to metastasize in vivo constitutively downregulated SNPH and exhibited higher oxidative stress, reduced cell proliferation, and increased cell motility. Therefore, SNPH is a stress-regulated mitochondrial switch of the cell proliferation-motility balance in cancer, and its pathway may represent a therapeutic target.
Collapse
Affiliation(s)
- M Cecilia Caino
- Prostate Cancer Discovery and Development Program.,Tumor Microenvironment and Metastasis Program, and
| | - Jae Ho Seo
- Prostate Cancer Discovery and Development Program.,Tumor Microenvironment and Metastasis Program, and
| | - Yuan Wang
- Prostate Cancer Discovery and Development Program.,Tumor Microenvironment and Metastasis Program, and
| | - Dayana B Rivadeneira
- Prostate Cancer Discovery and Development Program.,Tumor Microenvironment and Metastasis Program, and
| | - Dmitry I Gabrilovich
- Translational Tumor Immunology Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Eui Tae Kim
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Lucia R Languino
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program.,Tumor Microenvironment and Metastasis Program, and
| |
Collapse
|
35
|
Chen G, Fu Q, Yu F, Ren R, Liu Y, Cao Z, Li G, Zhao X, Chen L, Wang H, You J. Wide-Acidity-Range pH Fluorescence Probes for Evaluation of Acidification in Mitochondria and Digestive Tract Mucosa. Anal Chem 2017; 89:8509-8516. [DOI: 10.1021/acs.analchem.7b02164] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Guang Chen
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Key
Laboratory of Coastal Environmental Processes and Ecological Remediation,
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
- Shandong
Province Key Laboratory of Detection Technology for Tumor Makers,
College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Qiang Fu
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Fabiao Yu
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Key
Laboratory of Coastal Environmental Processes and Ecological Remediation,
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Rui Ren
- Shandong
Province Key Laboratory of Detection Technology for Tumor Makers,
College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yuxia Liu
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Ziping Cao
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Guoliang Li
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Xianen Zhao
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Lingxin Chen
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Key
Laboratory of Coastal Environmental Processes and Ecological Remediation,
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Hua Wang
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Jinmao You
- The
Key Laboratory of Life-Organic Analysis; Key Laboratory of Pharmaceutical
Intermediates and Analysis of Natural Medicine, College of Chemistry
and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Key
Laboratory of Coastal Environmental Processes and Ecological Remediation,
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| |
Collapse
|
36
|
Mori MP, Costa RAP, Soltys DT, Freire TDS, Rossato FA, Amigo I, Kowaltowski AJ, Vercesi AE, de Souza-Pinto NC. Lack of XPC leads to a shift between respiratory complexes I and II but sensitizes cells to mitochondrial stress. Sci Rep 2017; 7:155. [PMID: 28273955 PMCID: PMC5427820 DOI: 10.1038/s41598-017-00130-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 02/08/2017] [Indexed: 12/13/2022] Open
Abstract
Genomic instability drives tumorigenesis and DNA repair defects are associated with elevated cancer. Metabolic alterations are also observed during tumorigenesis, although a causal relationship between these has not been clearly established. Xeroderma pigmentosum (XP) is a DNA repair disease characterized by early cancer. Cells with reduced expression of the XPC protein display a metabolic shift from OXPHOS to glycolysis, which was linked to accumulation of nuclear DNA damage and oxidants generation via NOX-1. Using XP-C cells, we show that mitochondrial respiratory complex I (CI) is impaired in the absence of XPC, while complex II (CII) is upregulated in XP-C cells. The CI/CII metabolic shift was dependent on XPC, as XPC complementation reverted the phenotype. We demonstrate that mitochondria are the primary source of H2O2 and glutathione peroxidase activity is compromised. Moreover, mtDNA is irreversibly damaged and accumulates deletions. XP-C cells were more sensitive to the mitochondrial inhibitor antimycin A, an effect also prevented in XPC-corrected cells. Our results show that XPC deficiency leads to alterations in mitochondrial redox balance with a CI/CII shift as a possible adaptation to lower CI activity, but at the cost of sensitizing XP-C cells to mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Mateus P Mori
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Rute A P Costa
- Department of Clinical Pathology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Daniela T Soltys
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Thiago de S Freire
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Franco A Rossato
- Department of Clinical Pathology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ignácio Amigo
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Aníbal E Vercesi
- Department of Clinical Pathology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Nadja C de Souza-Pinto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
37
|
Cancer Cell Mitochondria Targeting by Pancratistatin Analogs is Dependent on Functional Complex II and III. Sci Rep 2017; 7:42957. [PMID: 28220885 PMCID: PMC5318952 DOI: 10.1038/srep42957] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/17/2017] [Indexed: 11/16/2022] Open
Abstract
Enhanced mitochondrial stability and decreased dependence on oxidative phosphorylation confer an acquired resistance to apoptosis in cancer cells, but may present opportunities for therapeutic intervention. The compound pancratistatin (PST) has been shown to selectively induce apoptosis in cancer cells. However, its low availability in nature has hindered its clinical advancement. We synthesized PST analogs and a medium-throughput screen was completed. Analogs SVTH-7, -6, and -5 demonstrated potent anti-cancer activity greater than PST and several standard chemotherapeutics. They disrupted mitochondrial function, activated the intrinsic apoptotic pathway, and reduced growth of tumor xenografts in vivo. Interestingly, the pro-apoptotic effects of SVTH-7 on cancer cells and mitochondria were abrogated with the inhibition of mitochondrial complex II and III, suggesting mitochondrial or metabolic vulnerabilities may be exploited by this analog. This work provides a scaffold for characterizing distinct mitochondrial and metabolic features of cancer cells and reveals several lead compounds with high therapeutic potential.
Collapse
|
38
|
Hardonnière K, Huc L, Sergent O, Holme JA, Lagadic-Gossmann D. Environmental carcinogenesis and pH homeostasis: Not only a matter of dysregulated metabolism. Semin Cancer Biol 2017; 43:49-65. [PMID: 28088583 DOI: 10.1016/j.semcancer.2017.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/18/2022]
Abstract
According to the World Health Organization, around 20% of all cancers would be due to environmental factors. Among these factors, several chemicals are indeed well recognized carcinogens. The widespread contaminant benzo[a]pyrene (B[a]P), an often used model carcinogen of the polycyclic aromatic hydrocarbons' family, has been suggested to target most, if not all, cancer hallmarks described by Hanahan and Weinberg. It is classified as a group I carcinogen by the International Agency for Research on Cancer; however, the precise intracellular mechanisms underlying its carcinogenic properties remain yet to be thoroughly defined. Recently, the pH homeostasis, a well known regulator of carcinogenic processes, was suggested to be a key actor in both cell death and Warburg-like metabolic reprogramming induced upon B[a]P exposure. The present review will highlight those data with the aim of favoring research on the role of H+ dynamics in environmental carcinogenesis.
Collapse
Affiliation(s)
- Kévin Hardonnière
- Institut national de la santé et de la recherche médicale (Inserm), Institut de recherche en santé, environnement et travail (Irset - Inserm UMR 1085), F-35043 Rennes, France; Université de Rennes 1, Structure fédérative de recherche Biosit, UMS CNRS 3480/US Inserm 018, F 35043 Rennes, France
| | - Laurence Huc
- INRA UMR 1331 ToxAlim (Research Center in Food Toxicology), University of Toulouse ENVT, INP, UPS, 180 Chemin de Tournefeuille, F-31027, France
| | - Odile Sergent
- Institut national de la santé et de la recherche médicale (Inserm), Institut de recherche en santé, environnement et travail (Irset - Inserm UMR 1085), F-35043 Rennes, France; Université de Rennes 1, Structure fédérative de recherche Biosit, UMS CNRS 3480/US Inserm 018, F 35043 Rennes, France
| | - Jørn A Holme
- Domain of Infection Control, Environment and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Dominique Lagadic-Gossmann
- Institut national de la santé et de la recherche médicale (Inserm), Institut de recherche en santé, environnement et travail (Irset - Inserm UMR 1085), F-35043 Rennes, France; Université de Rennes 1, Structure fédérative de recherche Biosit, UMS CNRS 3480/US Inserm 018, F 35043 Rennes, France.
| |
Collapse
|
39
|
Magcwebeba T, Swart P, Swanevelder S, Joubert E, Gelderblom W. Anti-Inflammatory Effects of Aspalathus linearis and Cyclopia spp. Extracts in a UVB/Keratinocyte (HaCaT) Model Utilising Interleukin-1α Accumulation as Biomarker. Molecules 2016; 21:molecules21101323. [PMID: 27706097 PMCID: PMC6274390 DOI: 10.3390/molecules21101323] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/16/2016] [Accepted: 09/26/2016] [Indexed: 01/06/2023] Open
Abstract
Ultraviolet B (UVB) radiation is one of the major predisposing risk factors of skin cancer. The anticancer and photoprotective effects of unoxidized rooibos (Aspalathus linearis) and honeybush (Cyclopia) herbal teas, containing high levels of dihydrochalones and xanthones, respectively, have been demonstrated in skin cancer models in vivo. In the current study, the anti-inflammatory effects of methanol and aqueous extracts of these herbal teas were investigated in a UVB/HaCaT keratinocyte model with intracellular interleukin-1α (icIL-1α) accumulation as a biomarker. Extracts of green tea (Camellia sinensis) served as benchmark. Both extracts of green tea and rooibos, as well as the aqueous extract of C. intermedia, enhanced UVB-induced inhibition of cell viability, proliferation and induction of apoptosis, facilitating the removal of icIL-1α. The underlying mechanisms may involve mitochondrial dysfunction exhibiting pro-oxidant responses via polyphenol-iron interactions. The methanol extracts of honeybush, however, protected against UVB-induced reduction of cell growth parameters, presumably via antioxidant mechanisms that prevented the removal of highly inflamed icIL-1α-containing keratinocytes via apoptosis. The dual antioxidant and/or pro-oxidant role of the polyphenolic herbal tea constituents should be considered in developing preventive strategies against UVB-induced skin carcinogenesis. The indirect removal of UVB damaged keratinocytes by herbal tea extracts via apoptosis may find application in the prevention of photo-induced inflammation.
Collapse
Affiliation(s)
- Tandeka Magcwebeba
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland (Stellenbosch) 7602, South Africa.
| | - Pieter Swart
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland (Stellenbosch) 7602, South Africa.
| | - Sonja Swanevelder
- Biostatistics Unit, South African Medical Research Council, P.O. Box 19070, Tygerberg 7505, South Africa.
| | - Elizabeth Joubert
- Post-Harvest and Wine Technology Division, Agricultural Research Council (Infruitec-Nietvoorbij), Private Bag X5026, Stellenbosch 7599, South Africa.
- Department of Food Science, Stellenbosch University, Private Bag X1, Matieland (Stellenbosch) 7602, South Africa.
| | - Wentzel Gelderblom
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland (Stellenbosch) 7602, South Africa.
- Institute of Biomedical and Microbial Biotechnology, Cape Peninsula University of Technology, P.O. Box 1906, Bellville 7535, South Africa.
| |
Collapse
|
40
|
Bezawork-Geleta A, Dong L, Rohlena J, Neuzil J. The Assembly Factor SDHAF2 Is Dispensable for Flavination of the Catalytic Subunit of Mitochondrial Complex II in Breast Cancer Cells. J Biol Chem 2016; 291:21414-21420. [PMID: 27587393 DOI: 10.1074/jbc.c116.755017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 08/27/2016] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial complex II or succinate dehydrogenase (SDH) is at the crossroads of oxidative phosphorylation and the tricarboxylic acid cycle. It has been shown that Sdh5 (SDHAF2/SDH5 in mammals) is required for flavination of the subunit Sdh1 (SDHA in human cells) in yeast. Here we demonstrate that in human breast cancer cells, SDHAF2/SDH5 is dispensable for SDHA flavination. In contrast to yeast, CRISPR-Cas9 nickase-mediated SDHAF2 KO breast cancer cells feature flavinated SDHA and retain fully assembled and functional complex II, as well as normal mitochondrial respiration. Our data show that SDHA flavination is independent of SDHAF2 in breast cancer cells, employing an alternative mechanism.
Collapse
Affiliation(s)
| | - Lanfeng Dong
- From the School of Medical Science, Griffith University, Southport, 4222 Queensland, Australia and
| | - Jakub Rohlena
- the Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Jiri Neuzil
- From the School of Medical Science, Griffith University, Southport, 4222 Queensland, Australia and .,the Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| |
Collapse
|
41
|
The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival. Sci Rep 2016; 6:30776. [PMID: 27488617 PMCID: PMC4973274 DOI: 10.1038/srep30776] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/10/2016] [Indexed: 12/30/2022] Open
Abstract
Cancer cells display alterations in many cellular processes. One core hallmark of cancer is the Warburg effect which is a glycolytic reprogramming that allows cells to survive and proliferate. Although the contributions of environmental contaminants to cancer development are widely accepted, the underlying mechanisms have to be clarified. Benzo[a]pyrene (B[a]P), the prototype of polycyclic aromatic hydrocarbons, exhibits genotoxic and carcinogenic effects, and it is a human carcinogen according to the International Agency for Research on Cancer. In addition to triggering apoptotic signals, B[a]P may induce survival signals, both of which are likely to be involved in cancer promotion. We previously suggested that B[a]P-induced mitochondrial dysfunctions, especially membrane hyperpolarization, might trigger cell survival signaling in rat hepatic epithelial F258 cells. Here, we further characterized these dysfunctions by focusing on energy metabolism. We found that B[a]P promoted a metabolic reprogramming. Cell respiration decreased and lactate production increased. These changes were associated with alterations in the tricarboxylic acid cycle which likely involve a dysfunction of the mitochondrial complex II. The glycolytic shift relied on activation of the Na+/H+ exchanger 1 (NHE1) and appeared to be a key feature in B[a]P-induced cell survival related to changes in cell phenotype (epithelial-to-mesenchymal transition and cell migration).
Collapse
|
42
|
Kruspig B, Valter K, Skender B, Zhivotovsky B, Gogvadze V. Targeting succinate:ubiquinone reductase potentiates the efficacy of anticancer therapy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2065-71. [DOI: 10.1016/j.bbamcr.2016.04.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/12/2016] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
|
43
|
Succinate, an intermediate in metabolism, signal transduction, ROS, hypoxia, and tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1086-1101. [PMID: 26971832 DOI: 10.1016/j.bbabio.2016.03.012] [Citation(s) in RCA: 330] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/06/2016] [Accepted: 03/07/2016] [Indexed: 12/31/2022]
Abstract
Succinate is an important metabolite at the cross-road of several metabolic pathways, also involved in the formation and elimination of reactive oxygen species. However, it is becoming increasingly apparent that its realm extends to epigenetics, tumorigenesis, signal transduction, endo- and paracrine modulation and inflammation. Here we review the pathways encompassing succinate as a metabolite or a signal and how these may interact in normal and pathological conditions.(1).
Collapse
|
44
|
Maio N, Ghezzi D, Verrigni D, Rizza T, Bertini E, Martinelli D, Zeviani M, Singh A, Carrozzo R, Rouault TA. Disease-Causing SDHAF1 Mutations Impair Transfer of Fe-S Clusters to SDHB. Cell Metab 2016; 23:292-302. [PMID: 26749241 PMCID: PMC4749439 DOI: 10.1016/j.cmet.2015.12.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/30/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
Abstract
SDHAF1 mutations cause a rare mitochondrial complex II (CII) deficiency, which manifests as infantile leukoencephalopathy with elevated levels of serum and white matter succinate and lactate. Here, we demonstrate that SDHAF1 contributes to iron-sulfur (Fe-S) cluster incorporation into the Fe-S subunit of CII, SDHB. SDHAF1 transiently binds to aromatic peptides of SDHB through an arginine-rich region in its C terminus and specifically engages a Fe-S donor complex, consisting of the scaffold, holo-ISCU, and the co-chaperone-chaperone pair, HSC20-HSPA9, through an LYR motif near its N-terminal domain. Pathogenic mutations of SDHAF1 abrogate binding to SDHB, which impairs biogenesis of holo-SDHB and results in LONP1-mediated degradation of SDHB. Riboflavin treatment was found to ameliorate the neurologic condition of patients. We demonstrate that riboflavin enhances flavinylation of SDHA and reduces levels of succinate and Hypoxia-Inducible Factor (HIF)-1α and -2α, explaining the favorable response of patients to riboflavin.
Collapse
Affiliation(s)
- Nunziata Maio
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 9000 Rockville Pike, 20892, Bethesda, MD, USA
| | - Daniele Ghezzi
- Unit of Molecular Neurogenetics, Foundation Carlo Besta Neurological Institute, Istituto di Ricovero e Cura a Carattere Scientifico, 20126 Milan, Italy
| | - Daniela Verrigni
- Unit for Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Teresa Rizza
- Unit for Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Enrico Bertini
- Unit for Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Diego Martinelli
- Unit of Metabolism, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Massimo Zeviani
- Mitochondrial Biology Unit, Medical Research Council, Hills Road, Cambridge CB2 0XY, UK
| | - Anamika Singh
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 9000 Rockville Pike, 20892, Bethesda, MD, USA
| | - Rosalba Carrozzo
- Unit for Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Tracey A Rouault
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 9000 Rockville Pike, 20892, Bethesda, MD, USA.
| |
Collapse
|
45
|
Song X, Hu M, Wang C, Xiao Y. Near-infrared fluorescent probes with higher quantum yields and neutral pKa values for the evaluation of intracellular pH. RSC Adv 2016. [DOI: 10.1039/c6ra11637j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Near-infrared fluorescent probes for pH, named pH-A and pH-B, for labeling cells to produce high resolution fluorescent images reflect the changes of intracellular pH.
Collapse
Affiliation(s)
- Xinbo Song
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116024
- China
| | - Mingyu Hu
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116024
- China
| | - Chao Wang
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116024
- China
| | - Yi Xiao
- State Key Laboratory of Fine Chemicals
- Dalian University of Technology
- Dalian 116024
- China
| |
Collapse
|
46
|
Kandola K, Bowman A, Birch-Machin MA. Oxidative stress - a key emerging impact factor in health, ageing, lifestyle and aesthetics. Int J Cosmet Sci 2015; 37 Suppl 2:1-8. [DOI: 10.1111/ics.12287] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/05/2015] [Indexed: 01/18/2023]
Affiliation(s)
- K. Kandola
- Dermatology; Medical School Newcastle University; Newcastle upon Tyne NE24HH U.K
| | - A. Bowman
- Dermatology; Medical School Newcastle University; Newcastle upon Tyne NE24HH U.K
| | - M. A. Birch-Machin
- Dermatology; Medical School Newcastle University; Newcastle upon Tyne NE24HH U.K
| |
Collapse
|
47
|
Guo L, Shestov AA, Worth AJ, Nath K, Nelson DS, Leeper DB, Glickson JD, Blair IA. Inhibition of Mitochondrial Complex II by the Anticancer Agent Lonidamine. J Biol Chem 2015; 291:42-57. [PMID: 26521302 PMCID: PMC4697178 DOI: 10.1074/jbc.m115.697516] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Indexed: 11/13/2022] Open
Abstract
The antitumor agent lonidamine (LND; 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylic acid) is known to interfere with energy-yielding processes in cancer cells. However, the effect of LND on central energy metabolism has never been fully characterized. In this study, we report that a significant amount of succinate is accumulated in LND-treated cells. LND inhibits the formation of fumarate and malate and suppresses succinate-induced respiration of isolated mitochondria. Utilizing biochemical assays, we determined that LND inhibits the succinate-ubiquinone reductase activity of respiratory complex II without fully blocking succinate dehydrogenase activity. LND also induces cellular reactive oxygen species through complex II, which reduced the viability of the DB-1 melanoma cell line. The ability of LND to promote cell death was potentiated by its suppression of the pentose phosphate pathway, which resulted in inhibition of NADPH and glutathione generation. Using stable isotope tracers in combination with isotopologue analysis, we showed that LND increased glutaminolysis but decreased reductive carboxylation of glutamine-derived α-ketoglutarate. Our findings on the previously uncharacterized effects of LND may provide potential combinational therapeutic approaches for targeting cancer metabolism.
Collapse
Affiliation(s)
- Lili Guo
- From the Penn Superfund Research and Training Program Center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics and
| | - Alexander A Shestov
- Laboratory of Molecular Imaging Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Andrew J Worth
- From the Penn Superfund Research and Training Program Center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics and
| | - Kavindra Nath
- Laboratory of Molecular Imaging Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - David S Nelson
- Laboratory of Molecular Imaging Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Dennis B Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Jerry D Glickson
- Laboratory of Molecular Imaging Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Ian A Blair
- From the Penn Superfund Research and Training Program Center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics and
| |
Collapse
|
48
|
Guzzo G, Sciacovelli M, Bernardi P, Rasola A. Inhibition of succinate dehydrogenase by the mitochondrial chaperone TRAP1 has anti-oxidant and anti-apoptotic effects on tumor cells. Oncotarget 2015; 5:11897-908. [PMID: 25564869 PMCID: PMC4323003 DOI: 10.18632/oncotarget.2472] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 09/13/2014] [Indexed: 12/20/2022] Open
Abstract
TRAP1 is a mitochondrial chaperone highly expressed in many tumor types; it inhibits respiratory complex II, down-modulating its succinate dehydrogenase (SDH) enzymatic activity. SDH inhibition in turn leads to a pseudohypoxic state caused by succinate-dependent HIF1α stabilization and promotes neoplastic growth. Here we report that TRAP1 inhibition of SDH also shields cells from oxidative insults and from the ensuing lethal opening of the mitochondrial permeability transition pore. This anti-oxidant activity of TRAP1 protects tumor cells from death in conditions of nutrient paucity that mimic those encountered in the neoplasm during the process of malignant accrual, and it is required for in vitro tumorigenic growth. Our findings demonstrate that SDH inhibition by TRAP1 is oncogenic not only by inducing pseudohypoxia, but also by protecting tumor cells from oxidative stress.
Collapse
Affiliation(s)
- Giulia Guzzo
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - Marco Sciacovelli
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy. Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Paolo Bernardi
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - Andrea Rasola
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| |
Collapse
|
49
|
Mitochondrial complex II is a source of the reserve respiratory capacity that is regulated by metabolic sensors and promotes cell survival. Cell Death Dis 2015. [PMID: 26225774 PMCID: PMC4650745 DOI: 10.1038/cddis.2015.202] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The survival of a cell depends on its ability to meet its energy requirements. We hypothesized that the mitochondrial reserve respiratory capacity (RRC) of a cell is a critical component of its bioenergetics that can be utilized during an increase in energy demand, thereby, enhancing viability. Our goal was to identify the elements that regulate and contribute to the development of RRC and its involvement in cell survival. The results show that activation of metabolic sensors, including pyruvate dehydrogenase and AMP-dependent kinase, increases cardiac myocyte RRC via a Sirt3-dependent mechanism. Notably, we identified mitochondrial complex II (cII) as a target of these metabolic sensors and the main source of RRC. Moreover, we show that RRC, via cII, correlates with enhanced cell survival after hypoxia. Thus, for the first time, we show that metabolic sensors via Sirt3 maximize the cellular RRC through activating cII, which enhances cell survival after hypoxia.
Collapse
|
50
|
Abstract
SIGNIFICANCE The molecular mechanism of aging is still vigorously debated, although a general consensus exists that mitochondria are significantly involved in this process. However, the previously postulated role of mitochondrial-derived reactive oxygen species (ROS) as the damaging agents inducing functional loss in aging has fallen out of favor in the recent past. In this review, we critically examine the role of ROS in aging in the light of recent advances on the relationship between mitochondrial structure and function. RECENT ADVANCES The functional mitochondrial respiratory chain is now recognized as a reflection of the dynamic association of respiratory complexes in the form of supercomplexes (SCs). Besides providing kinetic advantage (channeling), SCs control ROS generation by the respiratory chain, thus providing a means to regulate ROS levels in the cell. Depending on their concentration, these ROS are either physiological signals essential for the life of the cell or toxic species that damage cell structure and functions. CRITICAL ISSUES We propose that under physiological conditions the dynamic nature of SCs reversibly controls the generation of ROS as signals involved in mitochondrial-nuclear communication. During aging, there is a progressive loss of control of ROS generation so that their production is irreversibly enhanced, inducing a vicious circle in which signaling is altered and structural damage takes place. FUTURE DIRECTIONS A better understanding on the forces affecting SC association would allow the manipulation of ROS generation, directing these species to their physiological signaling role.
Collapse
Affiliation(s)
- Maria Luisa Genova
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| | - Giorgio Lenaz
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| |
Collapse
|