1
|
Choe M, Ekvik AE, Stalnaker G, Shin HR, Titov DV. Genetically encoded tool for manipulation of ΔΨm identifies its role as the driver of ISR induced by ATP synthase dysfunction. Cell Chem Biol 2025; 32:620-630.e6. [PMID: 40250406 PMCID: PMC12011318 DOI: 10.1016/j.chembiol.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/19/2024] [Accepted: 03/18/2025] [Indexed: 04/20/2025]
Abstract
Mitochondrial membrane potential (ΔΨm) is one of the key parameters controlling cellular bioenergetics. Investigation of the role of ΔΨm in live cells is complicated by a lack of tools for its direct manipulation without off-target effects. Here, we adopted the uncoupling protein UCP1 from brown adipocytes as a genetically encoded tool for direct manipulation of ΔΨm. We validated the ability of exogenously expressed UCP1 to induce uncoupled respiration and lower ΔΨm in mammalian cells. UCP1 expression lowered ΔΨm to the same extent as chemical uncouplers but did not inhibit cell proliferation, suggesting that it manipulates ΔΨm without the off-target effects of chemical uncouplers. Using UCP1, we revealed that elevated ΔΨm is the driver of the integrated stress response induced by ATP synthase inhibition in mammalian cells.
Collapse
Affiliation(s)
- Mangyu Choe
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alex E Ekvik
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gretchen Stalnaker
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hijai R Shin
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Denis V Titov
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
2
|
Islam MA, Sehar U, Sultana OF, Mukherjee U, Brownell M, Kshirsagar S, Reddy PH. SuperAgers and centenarians, dynamics of healthy ageing with cognitive resilience. Mech Ageing Dev 2024; 219:111936. [PMID: 38657874 DOI: 10.1016/j.mad.2024.111936] [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: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Graceful healthy ageing and extended longevity is the most desired goal for human race. The process of ageing is inevitable and has a profound impact on the gradual deterioration of our physiology and health since it triggers the onset of many chronic conditions like dementia, osteoporosis, diabetes, arthritis, cancer, and cardiovascular disease. However, some people who lived/live more than 100 years called 'Centenarians" and how do they achieve their extended lifespans are not completely understood. Studying these unknown factors of longevity is important not only to establish a longer human lifespan but also to manage and treat people with shortened lifespans suffering from age-related morbidities. Furthermore, older adults who maintain strong cognitive function are referred to as "SuperAgers" and may be resistant to risk factors linked to cognitive decline. Investigating the mechanisms underlying their cognitive resilience may contribute to the development of therapeutic strategies that support the preservation of cognitive function as people age. The key to a long, physically, and cognitively healthy life has been a mystery to scientists for ages. Developments in the medical sciences helps us to a better understanding of human physiological function and greater access to medical care has led us to an increase in life expectancy. Moreover, inheriting favorable genetic traits and adopting a healthy lifestyle play pivotal roles in promoting longer and healthier lives. Engaging in regular physical activity, maintaining a balanced diet, and avoiding harmful habits such as smoking contribute to overall well-being. The synergy between positive lifestyle choices, access to education, socio-economic factors, environmental determinants and genetic supremacy enhances the potential for a longer and healthier life. Our article aims to examine the factors associated with healthy ageing, particularly focusing on cognitive health in centenarians. We will also be discussing different aspects of ageing including genomic instability, metabolic burden, oxidative stress and inflammation, mitochondrial dysfunction, cellular senescence, immunosenescence, and sarcopenia.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Malcolm Brownell
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
3
|
Gao L, Peng L, Wang J, Zhang JH, Xia Y. Mitochondrial stress: a key role of neuroinflammation in stroke. J Neuroinflammation 2024; 21:44. [PMID: 38321473 PMCID: PMC10845693 DOI: 10.1186/s12974-024-03033-7] [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: 11/03/2023] [Accepted: 01/27/2024] [Indexed: 02/08/2024] Open
Abstract
Stroke is a clinical syndrome characterized by an acute, focal neurological deficit, primarily caused by the occlusion or rupture of cerebral blood vessels. In stroke, neuroinflammation emerges as a pivotal event contributing to neuronal cell death. The occurrence and progression of neuroinflammation entail intricate processes, prominently featuring mitochondrial dysfunction and adaptive responses. Mitochondria, a double membrane-bound organelle are recognized as the "energy workshop" of the body. Brain is particularly vulnerable to mitochondrial disturbances due to its high energy demands from mitochondria-related energy production. The interplay between mitochondria and neuroinflammation plays a significant role in the pathogenesis of stroke. The biological and pathological consequences resulting from mitochondrial stress have substantial implications for cerebral function. Mitochondrial stress serves as an adaptive mechanism aimed at mitigating the stress induced by the import of misfolded proteins, which occurs in response to stroke. This adaptive response involves a reduction in misfolded protein accumulation and overall protein synthesis. The influence of mitochondrial stress on the pathological state of stroke is underscored by its capacity to interact with neuroinflammation. The impact of mitochondrial stress on neuroinflammation varies according to its severity. Moderate mitochondrial stress can bolster cellular adaptive defenses, enabling cells to better withstand detrimental stressors. In contrast, sustained and excessive mitochondrial stress detrimentally affects cellular and tissue integrity. The relationship between neuroinflammation and mitochondrial stress depends on the degree of mitochondrial stress present. Understanding its role in stroke pathogenesis is instrumental in excavating the novel treatment of stroke. This review aims to provide the evaluation of the cross-talk between mitochondrial stress and neuroinflammation within the context of stroke. We aim to reveal how mitochondrial stress affects neuroinflammation environment in stroke.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Li Peng
- Department of Ophthalmology, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
| | - Jian Wang
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Neurosurgery and Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA.
| | - Ying Xia
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China.
| |
Collapse
|
4
|
Choe M, Titov DV. Genetically encoded tool for manipulation of ΔΨm identifies the latter as the driver of integrative stress response induced by ATP Synthase dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573435. [PMID: 38234735 PMCID: PMC10793441 DOI: 10.1101/2023.12.27.573435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Mitochondrial membrane potential (ΔΨm) is one of the key parameters controlling cellular bioenergetics. Investigation of the role of ΔΨm in live cells is complicated by a lack of tools for its direct manipulation without off-target effects. Here, we adopted the uncoupling protein UCP1 from brown adipocytes as a genetically encoded tool for direct manipulation of ΔΨm. We validated the ability of exogenously expressed UCP1 to induce uncoupled respiration and lower ΔΨm in mammalian cells. UCP1 expression lowered ΔΨm to the same extent as chemical uncouplers but did not inhibit cell proliferation, suggesting that it manipulates ΔΨm without the off-target effects of chemical uncouplers. Using UCP1, we revealed that elevated ΔΨm is the driver of the Integrated Stress Response induced by ATP synthase inhibition in mammalian cells.
Collapse
Affiliation(s)
- Mangyu Choe
- Department of Nutritional Sciences and Toxicology, University of California; Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California; Berkeley, CA 94720, USA
| | - Denis V Titov
- Department of Nutritional Sciences and Toxicology, University of California; Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California; Berkeley, CA 94720, USA
- Center for Computational Biology, University of California; Berkeley, CA 94720, USA
| |
Collapse
|
5
|
Abruscato G, Chiarelli R, Lazzara V, Punginelli D, Sugár S, Mauro M, Librizzi M, Di Stefano V, Arizza V, Vizzini A, Vazzana M, Luparello C. In Vitro Cytotoxic Effect of Aqueous Extracts from Leaves and Rhizomes of the Seagrass Posidonia oceanica (L.) Delile on HepG2 Liver Cancer Cells: Focus on Autophagy and Apoptosis. BIOLOGY 2023; 12:biology12040616. [PMID: 37106816 PMCID: PMC10135731 DOI: 10.3390/biology12040616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023]
Abstract
Aqueous extracts from Posidonia oceanica's green and brown (beached) leaves and rhizomes were prepared, submitted to phenolic compound and proteomic analysis, and examined for their potential cytotoxic effect on HepG2 liver cancer cells in culture. The chosen endpoints related to survival and death were cell viability and locomotory behavior, cell-cycle analysis, apoptosis and autophagy, mitochondrial membrane polarization, and cell redox state. Here, we show that 24 h exposure to both green-leaf- and rhizome-derived extracts decreased tumor cell number in a dose-response manner, with a mean half maximal inhibitory concentration (IC50) estimated at 83 and 11.5 μg of dry extract/mL, respectively. Exposure to the IC50 of the extracts appeared to inhibit cell motility and long-term cell replicating capacity, with a more pronounced effect exerted by the rhizome-derived preparation. The underlying death-promoting mechanisms identified involved the down-regulation of autophagy, the onset of apoptosis, the decrease in the generation of reactive oxygen species, and the dissipation of mitochondrial transmembrane potential, although, at the molecular level, the two extracts appeared to elicit partially differentiating effects, conceivably due to their diverse composition. In conclusion, P. oceanica extracts merit further investigation to develop novel promising prevention and/or treatment agents, as well as beneficial supplements for the formulation of functional foods and food-packaging material with antioxidant and anticancer properties.
Collapse
Affiliation(s)
- Giulia Abruscato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Roberto Chiarelli
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Valentina Lazzara
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Diletta Punginelli
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Simon Sugár
- MS Proteomics Research Group, Research Centre for Natural Sciences, Eötvös Loránd Research Network, 1117 Budapest, Hungary
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Mariangela Librizzi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Vita Di Stefano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128 Palermo, Italy
| |
Collapse
|
6
|
Murányi J, Duró C, Gurbi B, Móra I, Varga A, Németh K, Simon J, Csala M, Csámpai A. Novel Erlotinib-Chalcone Hybrids Diminish Resistance in Head and Neck Cancer by Inducing Multiple Cell Death Mechanisms. Int J Mol Sci 2023; 24:ijms24043456. [PMID: 36834866 PMCID: PMC9964293 DOI: 10.3390/ijms24043456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
In a search for novel therapeutic options for head and neck squamous cell carcinomas (HNSCCs) generally treated with limited therapeutic success, we synthesized a series of novel erlotinib-chalcone molecular hybrids with 1,2,3-triazole and alkyne linkers and evaluated them for their anticancer activity on Fadu, Detroit 562 and SCC-25 HNSCC cell lines. Time- and dose-dependent cell viability measurements disclosed a significantly increased efficiency of the hybrids compared to the 1:1 combination of erlotinib and a reference chalcone. The clonogenic assay demonstrated that hybrids eradicate HNSCC cells in low micromolar concentrations. Experiments focusing on potential molecular targets indicate that the hybrids trigger the anticancer effect by a complementary mechanism of action that is independent of the canonical targets of their molecular fragments. Confocal microscopic imaging and real-time apoptosis/necrosis detection assay pointed to slightly different cell death mechanisms induced by the most prominent triazole- and alkyne-tethered hybrids (6a and 13, respectively). While 6a featured the lowest IC50 values on each of the three HNSCC cell lines, in Detroit 562 cells, this hybrid induced necrosis more markedly compared to 13. The therapeutic potential indicated by the observed anticancer efficacy of our selected hybrid molecules validates the concept of development and justifies further investigation to reveal the underlying mechanism of action.
Collapse
Affiliation(s)
- József Murányi
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Cintia Duró
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
| | - Bianka Gurbi
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - István Móra
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Attila Varga
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Krisztina Németh
- MS Metabolomics Research Group, Centre for Structural Study, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok Krt. 2, H-1117 Budapest, Hungary
| | - József Simon
- Research Group of Analytical Chemistry, University of Pannonia, Egyetem utca 10, H-8200 Veszprém, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
- Correspondence: (M.C.); (A.C.)
| | - Antal Csámpai
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
- Correspondence: (M.C.); (A.C.)
| |
Collapse
|
7
|
Mohiuddin SG, Ghosh S, Kavousi P, Orman MA. Proton Motive Force Inhibitors Are Detrimental to Methicillin-Resistant Staphylococcus aureus Strains. Microbiol Spectr 2022; 10:e0202422. [PMID: 35943153 PMCID: PMC9430991 DOI: 10.1128/spectrum.02024-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) strains are tolerant of conventional antibiotics, making them extremely dangerous. Previous studies have shown the effectiveness of proton motive force (PMF) inhibitors at killing bacterial cells; however, whether these agents can launch a new treatment strategy to eliminate antibiotic-tolerant cells mandates further investigation. Here, using known PMF inhibitors and two different MRSA isolates, we showed that the bactericidal potency of PMF inhibitors seemed to correlate with their ability to disrupt PMF and permeabilize cell membranes. By screening a small chemical library to verify this correlation, we identified a subset of chemicals (including nordihydroguaiaretic acid, gossypol, trifluoperazine, and amitriptyline) that strongly disrupted PMF in MRSA cells by dissipating either the transmembrane electric potential (ΔΨ) or the proton gradient (ΔpH). These drugs robustly permeabilized cell membranes and reduced MRSA cell levels below the limit of detection. Overall, our study further highlights the importance of cellular PMF as a target for designing new bactericidal therapeutics for pathogens. IMPORTANCE Methicillin-resistant Staphylococcus aureus (MRSA) emerged as a major hypervirulent pathogen that causes severe health care-acquired infections. These pathogens can be multidrug-tolerant cells, which can facilitate the recurrence of chronic infections and the emergence of diverse antibiotic-resistant mutants. In this study, we aimed to investigate whether proton motive force (PMF) inhibitors can launch a new treatment strategy to eliminate MRSA cells. Our in-depth analysis showed that PMF inhibitors that strongly dissipate either the transmembrane electric potential or the proton gradient can robustly permeabilize cell membranes and reduce MRSA cell levels below the limit of detection.
Collapse
Affiliation(s)
- Sayed Golam Mohiuddin
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Sreyashi Ghosh
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Pouria Kavousi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Mehmet A. Orman
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
8
|
Trushina E, Trushin S, Hasan MF. Mitochondrial complex I as a therapeutic target for Alzheimer's disease. Acta Pharm Sin B 2022; 12:483-495. [PMID: 35256930 PMCID: PMC8897152 DOI: 10.1016/j.apsb.2021.11.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/01/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD), the most prominent form of dementia in the elderly, has no cure. Strategies focused on the reduction of amyloid beta or hyperphosphorylated Tau protein have largely failed in clinical trials. Novel therapeutic targets and strategies are urgently needed. Emerging data suggest that in response to environmental stress, mitochondria initiate an integrated stress response (ISR) shown to be beneficial for healthy aging and neuroprotection. Here, we review data that implicate mitochondrial electron transport complexes involved in oxidative phosphorylation as a hub for small molecule-targeted therapeutics that could induce beneficial mitochondrial ISR. Specifically, partial inhibition of mitochondrial complex I has been exploited as a novel strategy for multiple human conditions, including AD, with several small molecules being tested in clinical trials. We discuss current understanding of the molecular mechanisms involved in this counterintuitive approach. Since this strategy has also been shown to enhance health and life span, the development of safe and efficacious complex I inhibitors could promote healthy aging, delaying the onset of age-related neurodegenerative diseases.
Collapse
Key Words
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- AIDS, acquired immunodeficiency syndrome
- AMP, adenosine monophosphate
- AMPK, AMP-activated protein kinase
- APP/PS1, amyloid precursor protein/presenilin 1
- ATP, adenosine triphosphate
- Alzheimer's disease
- Aβ, amyloid beta
- BBB, blood‒brain barrier
- BDNF, brain-derived neurotrophic factor
- CP2, tricyclic pyrone compound two
- Complex I inhibitors
- ER, endoplasmic reticulum
- ETC, electron transport chain
- FADH2, flavin adenine dinucleotide
- FDG-PET, fluorodeoxyglucose-positron emission tomography
- GWAS, genome-wide association study
- HD, Huntington's disease
- HIF-1α, hypoxia induced factor 1 α
- Healthy aging
- ISR, integrated stress response
- Integrated stress response
- LTP, long term potentiation
- MCI, mild cognitive impairment
- MPTP, 1-methyl 4-phenyl-1,2,3,6-tetrahydropyridine
- Mitochondria
- Mitochondria signaling
- Mitochondria targeted therapeutics
- NAD+ and NADH, nicotinamide adenine dinucleotide
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NRF2, nuclear factor E2-related factor 2
- Neuroprotection
- OXPHOS, oxidative phosphorylation
- PD, Parkinson's disease
- PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- PMF, proton-motive force
- RNAi, RNA interference
- ROS, reactive oxygen species
- T2DM, type II diabetes mellitus
- TCA, the tricarboxylic acid cycle
- mtDNA, mitochondrial DNA
- mtUPR, mitochondrial unfolded protein response
- pTau, hyper-phosphorylated Tau protein
- ΔpH, proton gradient
- Δψm, mitochondrial membrane potential
Collapse
Affiliation(s)
- Eugenia Trushina
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Sergey Trushin
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Md Fayad Hasan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
9
|
Song Y, Villeneuve DL. AOP Report: Uncoupling of Oxidative Phosphorylation Leading to Growth Inhibition via Decreased Cell Proliferation. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:2959-2967. [PMID: 34416019 PMCID: PMC10620627 DOI: 10.1002/etc.5197] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 05/22/2023]
Abstract
This report describes a novel adverse outcome pathway (AOP) on uncoupling of oxidative phosphorylation (OXPHOS) leading to growth inhibition via decreased adenosine triphosphate (ATP) pool and cell proliferation (AOPWiki, AOP263). Oxidative phosphorylation is a major metabolic process that produces the primary form of energy (ATP) supporting various biological functions. Uncoupling of OXPHOS is a widely recognized mode of action of many chemicals and is known to affect growth via different biological processes. Capturing these events in an AOP can greatly facilitate mechanistic understanding and hazard assessment of OXPHOS uncouplers and growth regulators in eukaryotes. The four proposed key events in this AOP are intentionally generalized to cover a wide range of organisms and stressors. Three out of four events can be measured using in vitro high-throughput bioassays, whereas for most organisms, growth inhibition can also be measured in a high-throughput format using standard in vivo toxicity test protocols. The key events and key event relationships in this AOP are further assessed for weight of evidence using evolved Bradford-Hill considerations. The overall confidence levels range from moderate to high with only a few uncertainties and inconsistencies. The chemical applicability domain of the AOP mainly contains protonophores uncouplers, which can be predicated using the quantitative structure-activity relationship (QSAR) approach and validated using in vitro high-throughput bioassays. The biological domain of the AOP basically covers all eukaryotes. The AOP described in this report is part of a larger AOP network linking uncoupling of OXPHOS to growth inhibition, and is considered highly relevant and applicable to both human health and ecological risk assessments.
Collapse
Affiliation(s)
- You Song
- Norwegian Institute for Water Research, Oslo, Norway
| | - Daniel L. Villeneuve
- Great Lakes Toxicology and Ecology Division, US Environmental Protection Agency, Duluth, Minnesota, USA
| |
Collapse
|
10
|
Zhang H, Chen N, Deng Z, Mai Y, Deng L, Chen G, Li Y, Pan B, Zhong W. Suppression of ANT2 by miR-137 Inhibits Prostate Tumorigenesis. Front Genet 2021; 12:687236. [PMID: 34539732 PMCID: PMC8448070 DOI: 10.3389/fgene.2021.687236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is a serious disease that affects men’s health. To date, no effective and long-lasting treatment option for this condition is available in clinical practice. ANT2 is highly expressed in a variety of hormone-related cancers, but its relationship and regulatory mechanism with PCa are unclear. In this study, we found that ANT2 expression was significantly upregulated in PCa tissues relative to control samples. Genetic knockdown of ANT2 effectively inhibited, while overexpression promoted, proliferation, migration, and invasion of PCa cells. In addition, miR-137 expression was reduced in prostate cancer tissues relative to control tissues. We identified a regulatory site for miR-137 in the 3′-UTR of ANT2 mRNA; luciferase reporter assays indicated that ANT2 is a direct target gene for miR-137. Transfecting cells with miR-137 mimics and/or an ANT2-encoding plasmid revealed that ANT2 promotes proliferation, migration, and invasion of PCa, whereas co-expression of miR-137 mimics inhibited these behaviors. These observations suggest that miR-137 mimics inhibit development of PCa by antagonizing expression of ANT2. Furthermore, tumorigenic assays in nude mice showed that miR-137 inhibitors abolished the inhibitory effect of ANT2 knockdown on PCa tumor growth. Collectively, our findings suggest that ANT2, a target gene of miR-137, is intimately involved in development of PCa, providing new evidence for the mechanism underlying pathogenesis of PCa as well as new options for targeted therapy.
Collapse
Affiliation(s)
- Heyuan Zhang
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Nanhui Chen
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Zhihai Deng
- Department of Urology, Gaozhou People's Hospital, Gaozhou, China
| | - Yang Mai
- Department of Urology, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Limin Deng
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yutong Li
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bin Pan
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weifeng Zhong
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China.,Department of Urology, Guangzhou Twelfth People's Hospital, Guangzhou, China
| |
Collapse
|
11
|
Iaubasarova IR, Khailova LS, Nazarov PA, Rokitskaya TI, Silachev DN, Danilina TI, Plotnikov EY, Denisov SS, Kirsanov RS, Korshunova GA, Kotova EA, Zorov DB, Antonenko YN. Linking 7-Nitrobenzo-2-oxa-1,3-diazole (NBD) to Triphenylphosphonium Yields Mitochondria-Targeted Protonophore and Antibacterial Agent. BIOCHEMISTRY (MOSCOW) 2021; 85:1578-1590. [PMID: 33705296 DOI: 10.1134/s000629792012010x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Appending lipophilic cations to small molecules has been widely used to produce mitochondria-targeted compounds with specific activities. In this work, we obtained a series of derivatives of the well-known fluorescent dye 7-nitrobenzo-2-oxa-1,3-diazole (NBD). According to the previous data [Denisov et al. (2014) Bioelectrochemistry, 98, 30-38], alkyl derivatives of NBD can uncouple isolated mitochondria at concentration of tens of micromoles despite a high pKa value (~11) of the dissociating group. Here, a number of triphenylphosphonium (TPP) derivatives linked to NBD via hydrocarbon spacers of varying length (C5, C8, C10, and C12) were synthesized (mitoNBD analogues), which accumulated in the mitochondria in an energy-dependent manner. NBD-C10-TPP (C10-mitoNBD) acted as a protonophore in artificial lipid membranes (liposomes) and uncoupled isolated mitochondria at micromolar concentrations, while the derivative with a shorter linker (NBD-C5-TPP, or C5-mitoNBD) exhibited no such activities. In accordance with this data, C10-mitoNBD was significantly more efficient than C5-mitoNBD in suppressing the growth of Bacillus subtilis. C10-mitoNBD and C12-mitoNBD demonstrated the highest antibacterial activity among the investigated analogues. C10-mitoNBD also exhibited the neuroprotective effect in the rat model of traumatic brain injury.
Collapse
Affiliation(s)
- I R Iaubasarova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.,Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - L S Khailova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - P A Nazarov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - T I Rokitskaya
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - D N Silachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - T I Danilina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - E Y Plotnikov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - S S Denisov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.,Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, Maastricht, 6229 ER, The Netherlands
| | - R S Kirsanov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - G A Korshunova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - E A Kotova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - D B Zorov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Y N Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
12
|
Schmidt CA, McLaughlin KL, Boykov IN, Mojalagbe R, Ranganathan A, Buddo KA, Lin CT, Fisher-Wellman KH, Neufer PD. Aglycemic growth enhances carbohydrate metabolism and induces sensitivity to menadione in cultured tumor-derived cells. Cancer Metab 2021; 9:3. [PMID: 33468237 PMCID: PMC7816515 DOI: 10.1186/s40170-021-00241-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most prevalent form of liver malignancy and carries poor prognoses due to late presentation of symptoms. Treatment of late-stage HCC relies heavily on chemotherapeutics, many of which target cellular energy metabolism. A key platform for testing candidate chemotherapeutic compounds is the intrahepatic orthotopic xenograft (IOX) model in rodents. Translational efficacy from the IOX model to clinical use is limited (in part) by variation in the metabolic phenotypes of the tumor-derived cells that can be induced by selective adaptation to subculture conditions. Methods In this study, a detailed multilevel systems approach combining microscopy, respirometry, potentiometry, and extracellular flux analysis (EFA) was utilized to examine metabolic adaptations that occur under aglycemic growth media conditions in HCC-derived (HEPG2) cells. We hypothesized that aglycemic growth would result in adaptive “aerobic poise” characterized by enhanced capacity for oxidative phosphorylation over a range of physiological energetic demand states. Results Aglycemic growth did not invoke adaptive changes in mitochondrial content, network complexity, or intrinsic functional capacity/efficiency. In intact cells, aglycemic growth markedly enhanced fermentative glycolytic substrate-level phosphorylation during glucose refeeding and enhanced responsiveness of both fermentation and oxidative phosphorylation to stimulated energy demand. Additionally, aglycemic growth induced sensitivity of HEPG2 cells to the provitamin menadione at a 25-fold lower dose compared to control cells. Conclusions These findings indicate that growth media conditions have substantial effects on the energy metabolism of subcultured tumor-derived cells, which may have significant implications for chemotherapeutic sensitivity during incorporation in IOX testing panels. Additionally, the metabolic phenotyping approach used in this study provides a practical workflow that can be incorporated with IOX screening practices to aid in deciphering the metabolic underpinnings of chemotherapeutic drug sensitivity. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-021-00241-0.
Collapse
Affiliation(s)
- Cameron A Schmidt
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA.,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Kelsey L McLaughlin
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA.,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Ilya N Boykov
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA.,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Rafiq Mojalagbe
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA
| | | | - Katherine A Buddo
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA.,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Chien-Te Lin
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA.,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Kelsey H Fisher-Wellman
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA. .,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, Greenville, NC, USA. .,Dept. of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
13
|
Donoso-Bustamante V, Borrego EA, Schiaffino-Bustamante Y, Gutiérrez DA, Millas-Vargas JP, Fuentes-Retamal S, Correa P, Carrillo I, Aguilera RJ, Miranda D, Chávez-Báez I, Pulgar R, Urra FA, Varela-Ramírez A, Araya-Maturana R. An acylhydroquinone derivative produces OXPHOS uncoupling and sensitization to BH3 mimetic ABT-199 (Venetoclax) in human promyelocytic leukemia cells. Bioorg Chem 2020; 100:103935. [PMID: 32454391 DOI: 10.1016/j.bioorg.2020.103935] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022]
Abstract
Since cancer cells have different mitochondrial bioenergetic requirements than non-cancerous cells, therapeutic inhibition of its mitochondrial functionality continues to be an important target for anticancer drug discovery. In this study, a series of acylhydroquinones with different acyl-chain length, and their chlorinated derivatives, in the aromatic ring, synthesized by Fries rearrangement under microwave irradiation, were evaluated for their anticancer activity in two leukemia cell lines. Findings from the primary and secondary screening of the 18 acylhydroquinones, tested at 5 µM on acute promyelocytic leukemia HL-60 and acute lymphoblastic leukemia CEM cells lines, identified an acylchlorohydroquinone (12) with a highly selective anti-proliferative effect toward HL-60 cells. This compound induced S-phase arrest in the cell cycle progression of HL-60 cells with insignificant toxicity on leukemic CEM cells and non-cancerous Hs27 cells. In HL-60 leukemic cells, 12 triggered increased mitochondrial NADH oxidation, increased respiration in presence of oligomycin (state 4o), mitochondrial depolarization, and ROS production, suggesting an uncoupling of OXPHOS. This provoked a metabolic adaptation dependent on AMPK/ACC/autophagy axis, having the mitochondrial β-oxidation a pro-survival role since the combination of 12 and etomoxir, a carnitine palmitoyl-transferase (CPT) inhibitor promoted extensive HL-60 cell death. Finally, 12-induced metabolic stress sensitized to HL-60 cells to cell death by the FDA-approved anti-leukemic drug ABT-199, a BH3 mimetic. Therefore, our results suggest that acylchlorohydroquinone is a promising scaffold in anti-promyelocytic leukemia drug research.
Collapse
Affiliation(s)
- Viviana Donoso-Bustamante
- Instituto de Química de Recursos Naturales, Universidad de Talca, Chile; Programa de Investigación Asociativa en Cáncer Gástrico, Universidad de Talca, Chile
| | - Edgar A Borrego
- Border Biomedical Research Center, Department of Biological Sciences, The University of Texas at El Paso, USA
| | | | - Denisse A Gutiérrez
- Border Biomedical Research Center, Department of Biological Sciences, The University of Texas at El Paso, USA
| | - Juan Pablo Millas-Vargas
- Instituto de Química de Recursos Naturales, Universidad de Talca, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Sebastián Fuentes-Retamal
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Programa de Investigación Asociativa en Cáncer Gástrico, Universidad de Talca, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Pablo Correa
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Ileana Carrillo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Renato J Aguilera
- Border Biomedical Research Center, Department of Biological Sciences, The University of Texas at El Paso, USA
| | - Dante Miranda
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ignacio Chávez-Báez
- Network for Snake Venom Research and Drug Discovery, Santiago, Chile; Laboratorio de Genómica y Genética de Interacciones Biológicas, INTA-Universidad de Chile, Santiago, Chile
| | - Rodrigo Pulgar
- Network for Snake Venom Research and Drug Discovery, Santiago, Chile; Laboratorio de Genómica y Genética de Interacciones Biológicas, INTA-Universidad de Chile, Santiago, Chile
| | - Félix A Urra
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile.
| | - Armando Varela-Ramírez
- Border Biomedical Research Center, Department of Biological Sciences, The University of Texas at El Paso, USA.
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, Universidad de Talca, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile.
| |
Collapse
|
14
|
Zaharia M, Mihai M, Roman T, Zbancioc G, Pui A, Gradinaru RV, Logigan C, Drochioiu G. Unusual ferrite induced photohydrolysis of dinitrophenols to nonaromatic and nontoxic derivatives. J Photochem Photobiol A Chem 2020. [DOI: 10.1016/j.jphotochem.2020.112497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Geisler JG. 2,4 Dinitrophenol as Medicine. Cells 2019; 8:cells8030280. [PMID: 30909602 PMCID: PMC6468406 DOI: 10.3390/cells8030280] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022] Open
Abstract
In the sanctity of pure drug discovery, objective reasoning can become clouded when pursuing ideas that appear unorthodox, but are spot on physiologically. To put this into historical perspective, it was an unorthodox idea in the 1950’s to suggest that warfarin, a rat poison, could be repositioned into a breakthrough drug in humans to protect against strokes as a blood thinner. Yet it was approved in 1954 as Coumadin® and has been prescribed to billions of patients as a standard of care. Similarly, no one can forget the horrific effects of thalidomide, prescribed or available without a prescription, as both a sleeping pill and “morning sickness” anti-nausea medication targeting pregnant women in the 1950’s. The “thalidomide babies” became the case-in-point for the need of strict guidelines by the U.S. Food & Drug Administration (FDA) or full multi-species teratogenicity testing before drug approval. More recently it was found that thalidomide is useful in graft versus host disease, leprosy and resistant tuberculosis treatment, and as an anti-angiogenesis agent as a breakthrough drug for multiple myeloma (except for pregnant female patients). Decades of diabetes drug discovery research has historically focused on every possible angle, except, the energy-out side of the equation, namely, raising mitochondrial energy expenditure with chemical uncouplers. The idea of “social responsibility” allowed energy-in agents to be explored and the portfolio is robust with medicines of insulin sensitizers, insulin analogues, secretagogues, SGLT2 inhibitors, etc., but not energy-out medicines. The primary reason? It appeared unorthodox, to return to exploring a drug platform used in the 1930s in over 100,000 obese patients used for weight loss. This is over 80-years ago and prior to Dr Peter Mitchell explaining the mechanism of how mitochondrial uncouplers, like 2,4-dinitrophenol (DNP) even worked by three decades later in 1961. Although there is a clear application for metabolic disease, it was not until recently that this platform was explored for its merit at very low, weight-neutral doses, for treating insidious human illnesses and completely unrelated to weight reduction. It is known that mitochondrial uncouplers specifically target the entire organelle’s physiology non-genomically. It has been known for years that many neuromuscular and neurodegenerative diseases are associated with overt production of reactive oxygen species (ROSs), a rise in isoprostanes (biomarker of mitochondrial ROSs in urine or blood) and poor calcium (Ca2+) handing. It has also been known that mitochondrial uncouplers lower ROS production and Ca2+ overload. There is evidence that elevation of isoprostanes precedes disease onset, in Alzheimer’s Disease (AD). It is also curious, why so many neurodegenerative diseases of known and unknown etiology start at mid-life or later, such as Multiple Sclerosis (MS), Huntington Disease (HD), AD, Parkinson Disease, and Amyotrophic Lateral Sclerosis (ALS). Is there a relationship to a buildup of mutations that are sequestered over time due to ROSs exceeding the rate of repair? If ROS production were managed, could disease onset due to aging be delayed or prevented? Is it possible that most, if not all neurodegenerative diseases are manifested through mitochondrial dysfunction? Although DNP, a historic mitochondrial uncoupler, was used in the 1930s at high doses for obesity in well over 100,000 humans, and so far, it has never been an FDA-approved drug. This review will focus on the application of using DNP, but now, repositioned as a potential disease-modifying drug for a legion of insidious diseases at much lower and paradoxically, weight neutral doses. DNP will be addressed as a treatment for “metabesity”, an emerging term related to the global comorbidities associated with the over-nutritional phenotype; obesity, diabetes, nonalcoholic steatohepatitis (NASH), metabolic syndrome, cardiovascular disease, but including neurodegenerative disorders and accelerated aging. Some unexpected drug findings will be discussed, such as DNP’s induction of neurotrophic growth factors involved in neuronal heath, learning and cognition. For the first time in 80’s years, the FDA has granted (to Mitochon Pharmaceutical, Inc., Blue Bell, PA, USA) an open Investigational New Drug (IND) approval to begin rigorous clinical testing of DNP for safety and tolerability, including for the first ever, pharmacokinetic profiling in humans. Successful completion of Phase I clinical trial will open the door to explore the merits of DNP as a possible treatment of people with many truly unmet medical needs, including those suffering from HD, MS, PD, AD, ALS, Duchenne Muscular Dystrophy (DMD), and Traumatic Brain Injury (TBI).
Collapse
Affiliation(s)
- John G Geisler
- Mitochon Pharmaceuticals, Inc., 970 Cross Lane, Blue Bell, PA 19422, USA.
| |
Collapse
|
16
|
Vogt S, Ruppert V, Pankuweit S, Paletta JPJ, Rhiel A, Weber P, Irqsusi M, Cybulski P, Ramzan R. Myocardial insufficiency is related to reduced subunit 4 content of cytochrome c oxidase. J Cardiothorac Surg 2018; 13:95. [PMID: 30223867 PMCID: PMC6142347 DOI: 10.1186/s13019-018-0785-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/11/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Treatment of heart failure remains one of the most challenging task for intensive care medicine, cardiology and cardiac surgery. New options and better indicators are always required. Understanding the basic mechanisms underlying heart failure promote the development of adjusted therapy e.g. assist devices and monitoring of recovery. If cardiac failure is related to compromised cellular respiration of the heart, remains unclear. Myocardial respiration depends on Cytochrome c- Oxidase (CytOx) activity representing the rate limiting step for the mitochondrial respiratory chain. The enzymatic activity as well as mRNA expression of enzyme's mitochondrial encoded catalytic subunit 2, nuclear encoded regulatory subunit 4 and protein contents were studied in biopsies of cardiac patients suffering from myocardial insufficiency and dilated cardiomyopathy (DCM). METHODS Fifty-four patients were enrolled in the study and underwent coronary angiography. Thirty male patients (mean age: 45 +/- 15 yrs.) had a reduced ejection fraction (EF) 35 ± 12% below 45% and a left ventricular end diastolic diameter (LVEDD) of 71 ± 10 mm bigger than 56 mm. They were diagnosed as having idiopathic dilated cardiomyopathy (DCM) without coronary heart disease and NYHA-class 3 and 4. Additionally, 24 male patients (mean age: 52 +/- 11 yrs.) after exclusion of secondary cardiomyopathies, coronary artery or valve disease, served as control (EF: 68 ± 7, LVEDD: 51 ± 7 mm). Total RNA was extracted from two biopsies of each person. Real-time PCR analysis was performed with specific primers followed by a melt curve analysis. Corresponding protein expression in the tissue was studied with immune-histochemistry while enzymatic activity was evaluated by spectroscopy. RESULTS Gene and protein expression analysis of patients showed a significant decrease of subunit 4 (1.1 vs. 0.6, p < 0.001; 7.7 ± 3.1% vs. 2.8 ± 1.4%, p < 0.0001) but no differences in subunit 2. Correlations were found between reduced subunit 2 expression, low EF (r = 0.766, p < 0.00045) and increased LVEDD (r = 0.492, p < 0.0068). In case of DCM less subunit 4 expression and reduced shortening fraction (r = 0.524, p < 0.017) was found, but enzymatic activity was higher (0.08 ± 0.06 vs. 0.26 ± 0.08 U/mg, p < 0.001) although myocardial oxygen consumption continued to the same extent. CONCLUSION In case of myocardial insufficiency and DCM, decreased expression of COX 4 results in an impaired CytOx activity. Higher enzymatic activity but equal oxygen consumption contribute to the pathophysiology of the myocardial insufficiency and appears as an indicator of oxidative stress. This kind of dysregulation should be in the focus for the development of diagnostic and therapy procedures.
Collapse
Affiliation(s)
- Sebastian Vogt
- Cardiovascular Research Laboratories at the Biochemical Pharmacological Center, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany. .,Heart Surgery, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany.
| | - Volker Ruppert
- Department for Internal Medicine- Cardiology, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Sabine Pankuweit
- Department for Internal Medicine- Cardiology, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Jürgen P J Paletta
- Clinic for Orthopedics and Rheumatology, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Annika Rhiel
- Cardiovascular Research Laboratories at the Biochemical Pharmacological Center, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Petra Weber
- Cardiovascular Research Laboratories at the Biochemical Pharmacological Center, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Marc Irqsusi
- Heart Surgery, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Pia Cybulski
- Cardiovascular Research Laboratories at the Biochemical Pharmacological Center, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| | - Rabia Ramzan
- Cardiovascular Research Laboratories at the Biochemical Pharmacological Center, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany.,Heart Surgery, Philipps-University Marburg and Universitätsklinikum Gießen and Marburg GmbH, Marburg, Germany
| |
Collapse
|
17
|
Urra FA, Muñoz F, Córdova-Delgado M, Ramírez MP, Peña-Ahumada B, Rios M, Cruz P, Ahumada-Castro U, Bustos G, Silva-Pavez E, Pulgar R, Morales D, Varela D, Millas-Vargas JP, Retamal E, Ramírez-Rodríguez O, Pessoa-Mahana H, Pavani M, Ferreira J, Cárdenas C, Araya-Maturana R. FR58P1a; a new uncoupler of OXPHOS that inhibits migration in triple-negative breast cancer cells via Sirt1/AMPK/β1-integrin pathway. Sci Rep 2018; 8:13190. [PMID: 30181620 PMCID: PMC6123471 DOI: 10.1038/s41598-018-31367-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Highly malignant triple-negative breast cancer (TNBC) cells rely mostly on glycolysis to maintain cellular homeostasis; however, mitochondria are still required for migration and metastasis. Taking advantage of the metabolic flexibility of TNBC MDA-MB-231 cells to generate subpopulations with glycolytic or oxidative phenotypes, we screened phenolic compounds containing an ortho-carbonyl group with mitochondrial activity and identified a bromoalkyl-ester of hydroquinone named FR58P1a, as a mitochondrial metabolism-affecting compound that uncouples OXPHOS through a protonophoric mechanism. In contrast to well-known protonophore uncoupler FCCP, FR58P1a does not depolarize the plasma membrane and its effect on the mitochondrial membrane potential and bioenergetics is moderate suggesting a mild uncoupling of OXPHOS. FR58P1a activates AMPK in a Sirt1-dependent fashion. Although the activation of Sirt1/AMPK axis by FR58P1a has a cyto-protective role, selectively inhibits fibronectin-dependent adhesion and migration in TNBC cells but not in non-tumoral MCF10A cells by decreasing β1-integrin at the cell surface. Prolonged exposure to FR58P1a triggers a metabolic reprograming in TNBC cells characterized by down-regulation of OXPHOS-related genes that promote cell survival but comprise their ability to migrate. Taken together, our results show that TNBC cell migration is susceptible to mitochondrial alterations induced by small molecules as FR58P1a, which may have therapeutic implications.
Collapse
Affiliation(s)
- Félix A Urra
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
| | - Felipe Muñoz
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Miguel Córdova-Delgado
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - María Paz Ramírez
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Bárbara Peña-Ahumada
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Melany Rios
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Pablo Cruz
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Galdo Bustos
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Rodrigo Pulgar
- Laboratorio de Bioinformática y Expresión Génica, INTA-Universidad de Chile, El Líbano, 5524, Santiago, Chile
| | - Danna Morales
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
| | - Diego Varela
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
| | - Juan Pablo Millas-Vargas
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Evelyn Retamal
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Oney Ramírez-Rodríguez
- Campus Río Simpson, University of Aysén, Obispo Vielmo 62, Coyhaique, 5952122, Aysén, Chile
| | - Hernán Pessoa-Mahana
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Mario Pavani
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Casilla 7, Santiago, Chile
| | - Jorge Ferreira
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Casilla 7, Santiago, Chile
| | - César Cárdenas
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, 93106, United States.
- The Buck Institute for Research on Aging, Novato, CA, 94945, United States.
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales and Programa de Investigación Asociativa en Cáncer Gástrico, Universidad de Talca, casilla 747, Talca, Chile.
| |
Collapse
|
18
|
Weaver RJ, Koch RE, Hill GE. What maintains signal honesty in animal colour displays used in mate choice? Philos Trans R Soc Lond B Biol Sci 2018; 372:rstb.2016.0343. [PMID: 28533460 DOI: 10.1098/rstb.2016.0343] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2017] [Indexed: 12/15/2022] Open
Abstract
Many of the colour displays of animals are proposed to have evolved in response to female mate choice for honest signals of quality, but such honest signalling requires mechanisms to prevent cheating. The most widely accepted and cited mechanisms for ensuring signal honesty are based on the costly signalling hypothesis, which posits that costs associated with ornamentation prevent low-quality males from being highly ornamented. Alternatively, by the index hypothesis, honesty can be achieved via cost-free mechanisms if ornament production is causally linked to core physiological pathways. In this essay, we review how a costly signalling framework has shaped empirical research in mate choice for colourful male ornaments and emphasize that alternative interpretations are plausible under an index signalling framework. We discuss the challenges in both empirically testing and distinguishing between the two hypotheses, noting that they need not be mutually exclusive. Finally, we advocate for a comprehensive approach to studies of colour signals that includes the explicit consideration of cost-free mechanisms for honesty.This article is part of the themed issue 'Animal coloration: production, perception, function and application'.
Collapse
Affiliation(s)
- Ryan J Weaver
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Rebecca E Koch
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Geoffrey E Hill
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
19
|
Szelechowski M, Amoedo N, Obre E, Léger C, Allard L, Bonneu M, Claverol S, Lacombe D, Oliet S, Chevallier S, Le Masson G, Rossignol R. Metabolic Reprogramming in Amyotrophic Lateral Sclerosis. Sci Rep 2018; 8:3953. [PMID: 29500423 PMCID: PMC5834494 DOI: 10.1038/s41598-018-22318-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction in the spinal cord is a hallmark of amyotrophic lateral sclerosis (ALS), but the neurometabolic alterations during early stages of the disease remain unknown. Here, we investigated the bioenergetic and proteomic changes in ALS mouse motor neurons and patients' skin fibroblasts. We first observed that SODG93A mice presymptomatic motor neurons display alterations in the coupling efficiency of oxidative phosphorylation, along with fragmentation of the mitochondrial network. The proteome of presymptomatic ALS mice motor neurons also revealed a peculiar metabolic signature with upregulation of most energy-transducing enzymes, including the fatty acid oxidation (FAO) and the ketogenic components HADHA and ACAT2, respectively. Accordingly, FAO inhibition altered cell viability specifically in ALS mice motor neurons, while uncoupling protein 2 (UCP2) inhibition recovered cellular ATP levels and mitochondrial network morphology. These findings suggest a novel hypothesis of ALS bioenergetics linking FAO and UCP2. Lastly, we provide a unique set of data comparing the molecular alterations found in human ALS patients' skin fibroblasts and SODG93A mouse motor neurons, revealing conserved changes in protein translation, folding and assembly, tRNA aminoacylation and cell adhesion processes.
Collapse
Affiliation(s)
- M Szelechowski
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - N Amoedo
- Bordeaux University, 33000, Bordeaux, France
- INSERM U1211, MRGM, 33000, Bordeaux, France
| | - E Obre
- CELLOMET, Center of Functional Genomics (CGFB), 146 Rue Léo Saignat, 33000, Bordeaux, France
| | - C Léger
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - L Allard
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - M Bonneu
- Bordeaux University, 33000, Bordeaux, France
- Center of Functional Genomics (CGFB), Proteomic Facility, Bordeaux University, 33000, Bordeaux, France
| | - S Claverol
- Bordeaux University, 33000, Bordeaux, France
- Center of Functional Genomics (CGFB), Proteomic Facility, Bordeaux University, 33000, Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000, Bordeaux, France
- INSERM U1211, MRGM, 33000, Bordeaux, France
| | - S Oliet
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - S Chevallier
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - G Le Masson
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France.
- Bordeaux University, 33000, Bordeaux, France.
| | - R Rossignol
- Bordeaux University, 33000, Bordeaux, France.
- INSERM U1211, MRGM, 33000, Bordeaux, France.
| |
Collapse
|
20
|
Koch RE, Hill GE. Behavioural mating displays depend on mitochondrial function: a potential mechanism for linking behaviour to individual condition. Biol Rev Camb Philos Soc 2018; 93:1387-1398. [DOI: 10.1111/brv.12400] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/15/2018] [Accepted: 01/19/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Rebecca E. Koch
- Department of Biological Sciences; Auburn University; Auburn AL 36849 U.S.A
| | - Geoffrey E. Hill
- Department of Biological Sciences; Auburn University; Auburn AL 36849 U.S.A
| |
Collapse
|
21
|
Voituron Y, Josserand R, Le Galliard JF, Haussy C, Roussel D, Romestaing C, Meylan S. Chronic stress, energy transduction, and free-radical production in a reptile. Oecologia 2017; 185:195-203. [PMID: 28836018 DOI: 10.1007/s00442-017-3933-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/14/2017] [Indexed: 01/06/2023]
Abstract
Stress hormones, such as corticosterone, play a crucial role in orchestrating physiological reaction patterns shaping adapted responses to stressful environments. Concepts aiming at predicting individual and population responses to environmental stress typically consider that stress hormones and their effects on metabolic rate provide appropriate proxies for the energy budget. However, uncoupling between the biochemical processes of respiration, ATP production, and free-radical production in mitochondria may play a fundamental role in the stress response and associated life histories. In this study, we aim at dissecting sub-cellular mechanisms that link these three processes by investigating both whole-organism metabolism, liver mitochondrial oxidative phosphorylation processes (O2 consumption and ATP production) and ROS emission in Zootoca vivipara individuals exposed 21 days to corticosterone relative to a placebo. Corticosterone enhancement had no effect on mitochondrial activity and efficiency. In parallel, the corticosterone treatment increased liver mass and mitochondrial protein content suggesting a higher liver ATP production. We also found a negative correlation between mitochondrial ROS emission and plasma corticosterone level. These results provide a proximal explanation for enhanced survival after chronic exposure to corticosterone in this species. Importantly, none of these modifications affected resting whole-body metabolic rate. Oxygen consumption, ATP, and ROS emission were thus independently affected in responses to corticosterone increase suggesting that concepts and models aiming at linking environmental stress and individual responses may misestimate energy allocation possibilities.
Collapse
Affiliation(s)
- Yann Voituron
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés (U.M.R. CNRS 5023), Université Claude Bernard Lyon1, Université de Lyon, Bd du 11 novembre 1918, Bât. Darwin C, 69622, Villeurbanne Cedex, France.
| | - Rémy Josserand
- Institut d'Ecologie et des Sciences, de l'Environnement de Paris (iEES Paris)-UPMC-CNRS, Bat. A, 7ème étage cc237, quai Saint Bernard, 75252, Paris Cedex 05, France
| | - Jean-François Le Galliard
- Institut d'Ecologie et des Sciences, de l'Environnement de Paris (iEES Paris)-UPMC-CNRS, Bat. A, 7ème étage cc237, quai Saint Bernard, 75252, Paris Cedex 05, France
- Ecole Normale Supérieure, PSL Research University, CNRS, Centre de recherche en écologie expérimentale et prédictive (CEREEP-Ecotron IleDeFrance), UMS 3194, 78 rue du château, 77140, Saint-Pierre-Lès-Nemours, France
| | - Claudy Haussy
- Institut d'Ecologie et des Sciences, de l'Environnement de Paris (iEES Paris)-UPMC-CNRS, Bat. A, 7ème étage cc237, quai Saint Bernard, 75252, Paris Cedex 05, France
| | - Damien Roussel
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés (U.M.R. CNRS 5023), Université Claude Bernard Lyon1, Université de Lyon, Bd du 11 novembre 1918, Bât. Darwin C, 69622, Villeurbanne Cedex, France
| | - Caroline Romestaing
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés (U.M.R. CNRS 5023), Université Claude Bernard Lyon1, Université de Lyon, Bd du 11 novembre 1918, Bât. Darwin C, 69622, Villeurbanne Cedex, France
| | - Sandrine Meylan
- Institut d'Ecologie et des Sciences, de l'Environnement de Paris (iEES Paris)-UPMC-CNRS, Bat. A, 7ème étage cc237, quai Saint Bernard, 75252, Paris Cedex 05, France
- ESPE de Paris, Université Sorbonne Paris IV, 10 rue Molitor, 75016, Paris, France
| |
Collapse
|
22
|
Wang R, MoYung KC, Zhang MH, Poon K. UCP2- and non-UCP2-mediated electric current in eukaryotic cells exhibits different properties. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:19618-19631. [PMID: 26276275 DOI: 10.1007/s11356-015-5155-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 08/04/2015] [Indexed: 06/04/2023]
Abstract
Using live eukaryotic cells, including cancer cells, MCF-7 and HCT-116, normal hepatocytes and red blood cells in anode and potassium ferricyanide in cathode of MFC could generate bio-based electric current. Electrons and protons generated from the metabolic reaction in both cytosol and mitochondria contributing to the leaking would mediate the generation of electric current. Both resveratrol (RVT) and 2,4-dinitrophenol (DNP) used to induce proton leak in mitochondria were found to promote electric current production in all cells except red blood cells without mitochondria. Proton leak might be important for electric current production by bringing the charge balance in cells to enhance the further electron leak. The induced electric current by RVT can be blocked by Genipin, an inhibitor of UCP2-mediated proton leak, while that induced by DNP cannot. RVT could reduce reactive oxygen species (ROS) level in cells better than that of DNP. In addition, RVT increased mitochondrial membrane potential (MMP), while DNP decreased it. Results highly suggested the existence of at least two types of electric current that showed different properties. They included UCP2-mediated and non-UCP2-mediated electric current. UCP2-mediated electric current exhibited higher reactive oxygen species (ROS) reduction effect per unit electric current production than that of non-UCP2-mediated electric current. Higher UCP2-mediated electric current observed in cancer cells might contribute to the mechanism of drug resistence. Correlation could not be established between electric current production with either ROS and MMP without distinguishing the types of electric current.
Collapse
Affiliation(s)
- Ruihua Wang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, 6600 Nanfeng Road, Fengxian District, Shanghai, China, 201499.
| | - K C MoYung
- Program of Food Science and Technology, Division of Science and Technology, BNU-HKBU United International College, 28 Jinfeng Road, Tangjiawan, Zhuhai, Guangdong, China, 519085.
| | - M H Zhang
- Program of Food Science and Technology, Division of Science and Technology, BNU-HKBU United International College, 28 Jinfeng Road, Tangjiawan, Zhuhai, Guangdong, China, 519085
| | - Karen Poon
- Program of Food Science and Technology, Division of Science and Technology, BNU-HKBU United International College, 28 Jinfeng Road, Tangjiawan, Zhuhai, Guangdong, China, 519085.
| |
Collapse
|
23
|
Perspectives on mitochondrial uncoupling proteins-mediated neuroprotection. J Bioenerg Biomembr 2014; 47:119-31. [PMID: 25217852 DOI: 10.1007/s10863-014-9580-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
The integrity of mitochondrial function is essential to cell life. It follows that disturbances of mitochondrial function will lead to disruption of cell function, expressed as disease or even death. Considering that neuronal uncoupling proteins (UCPs) decrease reactive oxygen species (ROS) production at the expense of energy production, it is important to understand the underlying mechanisms by which UCPs control the balance between the production of adenosine triphosphate (ATP) and ROS in the context of normal physiological activity and in pathological conditions. Here we review the current understanding of neuronal UCPs-mediated respiratory uncoupling process by performing a survey in their physiology and regulation. The latest findings regarding neuronal UCPs physiological roles and their involvement and interest as potential targets for therapeutic intervention in brain diseases will also be exploited.
Collapse
|
24
|
Laafi J, Homedan C, Jacques C, Gueguen N, Schmitt C, Puy H, Reynier P, Carmen Martinez M, Malthièry Y. Pro-oxidant effect of ALA is implicated in mitochondrial dysfunction of HepG2 cells. Biochimie 2014; 106:157-66. [PMID: 25220386 DOI: 10.1016/j.biochi.2014.08.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 08/22/2014] [Indexed: 12/16/2022]
Abstract
Heme biosynthesis begins in the mitochondrion with the formation of delta-aminolevulinic acid (ALA). In acute intermittent porphyria, hereditary tyrosinemia type I and lead poisoning patients, ALA is accumulated in plasma and in organs, especially the liver. These diseases are also associated with neuromuscular dysfunction and increased incidence of hepatocellular carcinoma. Many studies suggest that this damage may originate from ALA-induced oxidative stress following its accumulation. Using the MnSOD as an oxidative stress marker, we showed here that ALA treatment of cultured cells induced ROS production, increasing with ALA concentration. The mitochondrial energetic function of ALA-treated HepG2 cells was further explored. Mitochondrial respiration and ATP content were reduced compared to control cells. For the 300 μM treatment, ALA induced a mitochondrial mass decrease and a mitochondrial network imbalance although neither necrosis nor apoptosis were observed. The up regulation of PGC-1, Tfam and ND5 genes was also found; these genes encode mitochondrial proteins involved in mitochondrial biogenesis activation and OXPHOS function. We propose that ALA may constitute an internal bioenergetic signal, which initiates a coordinated upregulation of respiratory genes, which ultimately drives mitochondrial metabolic adaptation within cells. The addition of an antioxidant, Manganese(III) tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP), resulted in improvement of maximal respiratory chain capacity with 300 μM ALA. Our results suggest that mitochondria, an ALA-production site, are more sensitive to pro-oxidant effect of ALA, and may be directly involved in pathophysiology of patients with inherited or acquired porphyria.
Collapse
Affiliation(s)
- Jihane Laafi
- LUNAM Université, INSERM UMR 1063, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| | - Chadi Homedan
- LUNAM Université, INSERM UMR 1063, IBIS, IRIS, rue des capucins, 49100 Angers, France; Centre Hospitalier Universitaire, Département de Biochimie et Génétique, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| | - Caroline Jacques
- LUNAM Université, INSERM UMR 1063, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| | - Naig Gueguen
- Centre Hospitalier Universitaire, Département de Biochimie et Génétique, IBIS, IRIS, rue des capucins, 49100 Angers, France; CNRS UMR 6214 - INSERM 1083, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| | - Caroline Schmitt
- Assistance Publique Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Université Paris Diderot, 178 rue des Renouillers, 92700 Colombes, France; INSERM U773, Centre de Recherche Biomédicale Bichat-Beaujon, Paris, France; Université Paris Diderot, 5 Rue Thomas Mann, 75013 Paris, France.
| | - Hervé Puy
- Assistance Publique Hôpitaux de Paris, Centre Français des Porphyries, Hôpital Louis Mourier, Université Paris Diderot, 178 rue des Renouillers, 92700 Colombes, France; INSERM U773, Centre de Recherche Biomédicale Bichat-Beaujon, Paris, France; Université Paris Diderot, 5 Rue Thomas Mann, 75013 Paris, France.
| | - Pascal Reynier
- Centre Hospitalier Universitaire, Département de Biochimie et Génétique, IBIS, IRIS, rue des capucins, 49100 Angers, France; CNRS UMR 6214 - INSERM 1083, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| | - Maria Carmen Martinez
- LUNAM Université, INSERM UMR 1063, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| | - Yves Malthièry
- LUNAM Université, INSERM UMR 1063, IBIS, IRIS, rue des capucins, 49100 Angers, France; Centre Hospitalier Universitaire, Département de Biochimie et Génétique, IBIS, IRIS, rue des capucins, 49100 Angers, France.
| |
Collapse
|
25
|
Abstract
In cell culture, extracellular guanosine increases extracellular adenosine by attenuating the disposition of extracellular adenosine (American Journal of Physiology – Cell Physiology 304: C406–C421, 2013). The goal of this investigation was to determine whether this “guanosine–adenosine mechanism” is operative in an intact organ. Twenty‐seven isolated, perfused mouse kidneys were subjected to metabolic poisons (iodoacetate plus 2,4‐dinitrophenol) to cause energy depletion and thereby stimulate renal adenosine production. Adenosine levels in the renal venous perfusate increased from a baseline of 36 ± 8 to 499 ± 96, 258 ± 50, and 71 ± 13 nmol/L at 15, 30, and 60 min, respectively, after administering metabolic poisons (% of basal; 1366 ± 229, 715 ± 128, and 206 ± 33, respectively). Changes in renal venous levels of guanosine closely mirrored the time course of changes in adenosine: baseline of 15 ± 2 to 157 ± 13, 121 ± 8, and 50 ± 5 nmol/L at 15, 30, and 60 min, respectively (% of basal; 1132 ± 104, 871 ± 59, and 400 ± 51, respectively). Freeze‐clamp experiments in 12 kidneys confirmed that metabolic poisons increased kidney tissue levels of adenosine and guanosine. In eight additional kidneys, we examined the ability of guanosine to reduce the renal clearance of exogenous adenosine; and these experiments revealed that guanosine significantly decreased the renal extraction of adenosine. Because guanosine is metabolized by purine nucleoside phosphorylase (PNPase), in another set of 16 kidneys we examined the effects of 8‐aminoguanine (PNPase inhibitor) on renal venous levels of adenosine and inosine (adenosine metabolite). Kidneys treated with 8‐aminoguanine showed a more robust increase in both adenosine and inosine in response to metabolic poisons. We conclude that in the intact kidney, guanosine regulates adenosine levels. In cell culture, extracellular guanosine increases extracellular adenosine by attenuating the disposition of extracellular adenosine (American Journal of Physiology – Cell Physiology 304: C406–C421, 2013). The goal of this study was to determine whether the “guanosine–adenosine mechanism” is operative in an intact organ. In isolated, perfused mouse kidneys, inhibition of energy production induced changes in renal venous levels of guanosine that closely mirrored the time course of changes in adenosine, and freeze‐clamp experiments confirmed that metabolic poisons similarly increased kidney tissue levels of adenosine and guanosine. Moreover, exogenous guanosine significantly decreased the renal extraction of exogenous adenosine, and inhibition of purine nucleoside phosphorylase (metabolizes guanosine) augmented the effects of energy depletion on renal levels of both guanosine and adenosine. We conclude that in the intact kidney, guanosine regulates adenosine levels.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Delbert G Gillespie
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
26
|
Shoda LKM, Woodhead JL, Siler SQ, Watkins PB, Howell BA. Linking physiology to toxicity using DILIsym®, a mechanistic mathematical model of drug-induced liver injury. Biopharm Drug Dispos 2013; 35:33-49. [DOI: 10.1002/bdd.1878] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/10/2013] [Accepted: 11/01/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Lisl K. M. Shoda
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Jeffrey L. Woodhead
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Scott Q. Siler
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Paul B. Watkins
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Brett A. Howell
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| |
Collapse
|
27
|
Plasmatic concentration of organochlorine lindane acts as metabolic disruptors in HepG2 liver cell line by inducing mitochondrial disorder. Toxicol Appl Pharmacol 2013; 272:325-34. [DOI: 10.1016/j.taap.2013.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 06/04/2013] [Accepted: 06/06/2013] [Indexed: 12/18/2022]
|
28
|
Attene-Ramos MS, Huang R, Sakamuru S, Witt KL, Beeson GC, Shou L, Schnellmann RG, Beeson CC, Tice RR, Austin CP, Xia M. Systematic study of mitochondrial toxicity of environmental chemicals using quantitative high throughput screening. Chem Res Toxicol 2013; 26:1323-32. [PMID: 23895456 DOI: 10.1021/tx4001754] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A goal of the Tox21 program is to transit toxicity testing from traditional in vivo models to in vitro assays that assess how chemicals affect cellular responses and toxicity pathways. A critical contribution of the NIH Chemical Genomics center (NCGC) to the Tox21 program is the implementation of a quantitative high throughput screening (qHTS) approach, using cell- and biochemical-based assays to generate toxicological profiles for thousands of environmental compounds. Here, we evaluated the effect of chemical compounds on mitochondrial membrane potential in HepG2 cells by screening a library of 1,408 compounds provided by the National Toxicology Program (NTP) in a qHTS platform. Compounds were screened over 14 concentrations, and results showed that 91 and 88 compounds disrupted mitochondrial membrane potential after treatment for 1 or 5 h, respectively. Seventy-six compounds active at both time points were clustered by structural similarity, producing 11 clusters and 23 singletons. Thirty-eight compounds covering most of the active chemical space were more extensively evaluated. Thirty-six of the 38 compounds were confirmed to disrupt mitochondrial membrane potential using a fluorescence plate reader, and 35 were confirmed using a high content imaging approach. Among the 38 compounds, 4 and 6 induced LDH release, a measure of cytotoxicity, at 1 or 5 h, respectively. Compounds were further assessed for mechanism of action (MOA) by measuring changes in oxygen consumption rate, which enabled the identification of 20 compounds as uncouplers. This comprehensive approach allows for the evaluation of thousands of environmental chemicals for mitochondrial toxicity and identification of possible MOAs.
Collapse
Affiliation(s)
- Matias S Attene-Ramos
- National Center for Advancing Translational Sciences, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mitochondria-targeted antioxidants and metabolic modulators as pharmacological interventions to slow ageing. Biotechnol Adv 2012; 31:563-92. [PMID: 23022622 DOI: 10.1016/j.biotechadv.2012.09.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 02/07/2023]
Abstract
Populations in many nations today are rapidly ageing. This unprecedented demographic change represents one of the main challenges of our time. A defining property of the ageing process is a marked increase in the risk of mortality and morbidity with age. The incidence of cancer, cardiovascular and neurodegenerative diseases increases non-linearly, sometimes exponentially with age. One of the most important tasks in biogerontology is to develop interventions leading to an increase in healthy lifespan (health span), and a better understanding of basic mechanisms underlying the ageing process itself may lead to interventions able to delay or prevent many or even all age-dependent conditions. One of the putative basic mechanisms of ageing is age-dependent mitochondrial deterioration, closely associated with damage mediated by reactive oxygen species (ROS). Given the central role that mitochondria and mitochondrial dysfunction play not only in ageing but also in apoptosis, cancer, neurodegeneration and other age-related diseases there is great interest in approaches to protect mitochondria from ROS-mediated damage. In this review, we explore strategies of targeting mitochondria to reduce mitochondrial oxidative damage with the aim of preventing or delaying age-dependent decline in mitochondrial function and some of the resulting pathologies. We discuss mitochondria-targeted and -localized antioxidants (e.g.: MitoQ, SkQ, ergothioneine), mitochondrial metabolic modulators (e.g. dichloroacetic acid), and uncouplers (e.g.: uncoupling proteins, dinitrophenol) as well as some alternative future approaches for targeting compounds to the mitochondria, including advances from nanotechnology.
Collapse
|
30
|
Vaughan RA, Garcia-Smith R, Bisoffi M, Trujillo KA, Conn CA. Effects of caffeine on metabolism and mitochondria biogenesis in rhabdomyosarcoma cells compared with 2,4-dinitrophenol. Nutr Metab Insights 2012; 5:59-70. [PMID: 23882149 PMCID: PMC3698477 DOI: 10.4137/nmi.s10233] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Purpose: This work investigated if treatment with caffeine or 2,4-dinitrophenol (DNP) induce expression of peroxisome proliferator-activated receptor coactivator 1 alpha (PGC-1α) and increase both mitochondrial biosynthesis and metabolism in skeletal muscle. Methods: Human rhabdomyosarcoma cells were treated with either ethanol control (0.1% final concentration) caffeine, or DNP at 250 or 500 μM for 16 or 24 hours. PGC-1α RNA levels were determined using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). PGC-1α protein and mitochondrial content was determined using flow cytometry and immunohistochemistry. Metabolism was determined by quantification of extracellular acidification rate and oxygen consumption rate. Results: Treatment with either caffeine or DNP induced PGC-1α RNA and protein as well as mitochondrial content compared with control. Treatment with caffeine and DNP also significantly increased oxidative metabolism and total metabolic rate compared with control. Caffeine similarly increased metabolism and mitochondrial content compared with DNP. Conclusion: This work identified that both caffeine and DNP significantly induce PGC-1α, and increase both metabolism and mitochondrial content in skeletal muscle.
Collapse
Affiliation(s)
- Roger A Vaughan
- Department of Health, Exercise and Sports Science, University of New Mexico, Albuquerque
| | | | | | | | | |
Collapse
|
31
|
Riedel A, Pignitter M, Hochkogler CM, Rohm B, Walker J, Bytof G, Lantz I, Somoza V. Caffeine dose-dependently induces thermogenesis but restores ATP in HepG2 cells in culture. Food Funct 2012; 3:955-64. [PMID: 22710994 DOI: 10.1039/c2fo30053b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Caffeine has been hypothesised as a thermogenic agent that might help to maintain a healthy body weight. Since very little is known about its actions on cellular energy metabolism, we investigated the effect of caffeine on mitochondrial oxidative phosphorylation, cellular energy supply and thermogenesis in HepG2 cells, and studied its action on fatty acid uptake and lipid accumulation in 3T3-L1 adipocytes at concentrations ranging from 30-1500 μM. In HepG2 cells, caffeine induced a depolarisation of the inner mitochondrial membrane, a feature of mitochondrial thermogenesis, both directly and after 24 h incubation. Increased concentrations of uncoupling protein-2 (UCP-2) also indicated a thermogenic activity of caffeine. Energy generating pathways, such as mitochondrial respiration, fatty acid oxidation and anaerobic lactate production, were attenuated by caffeine treatment. Nevertheless, HepG2 cells demonstrated a higher energy charge potential after exposure to caffeine that might result from energy restoration through attenuation of energy consuming pathways, as typically found in hibernating animals. In 3T3-L1 cells, in contrast, caffeine increased fatty acid uptake, but did not affect lipid accumulation. We provide evidence that caffeine stimulates thermogenesis but concomitantly causes energy restoration that may compensate enhanced energy expenditure.
Collapse
Affiliation(s)
- Annett Riedel
- Department of Nutritional and Physiological Chemistry, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
32
|
l-Lactate metabolism in HEP G2 cell mitochondria due to the l-lactate dehydrogenase determines the occurrence of the lactate/pyruvate shuttle and the appearance of oxaloacetate, malate and citrate outside mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1817:1679-90. [PMID: 22659615 DOI: 10.1016/j.bbabio.2012.05.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 05/22/2012] [Accepted: 05/24/2012] [Indexed: 01/03/2023]
Abstract
As part of an ongoing study of l-lactate metabolism both in normal and in cancer cells, we investigated whether and how l-lactate metabolism occurs in mitochondria of human hepatocellular carcinoma (Hep G2) cells. We found that Hep G2 cell mitochondria (Hep G2-M) possess an l-lactate dehydrogenase (ml-LDH) restricted to the inner mitochondrial compartments as shown by immunological analysis, confocal microscopy and by assaying ml-LDH activity in solubilized mitochondria. Cytosolic and mitochondrial l-LDHs were found to differ from one another in their saturation kinetics. Having shown that l-lactate itself can enter Hep G2 cells, we found that Hep G2-M swell in ammonium l-lactate, but not in ammonium pyruvate solutions, in a manner inhibited by mersalyl, this showing the occurrence of a carrier-mediated l-lactate transport in these mitochondria. Occurrence of the l-lactate/pyruvate shuttle and the appearance outside mitochondria of oxaloacetate, malate and citrate arising from l-lactate uptake and metabolism together with the low oxygen consumption and membrane potential generation are in favor of an anaplerotic role for l-LAC in Hep G2-M.
Collapse
|
33
|
Salin K, Luquet E, Rey B, Roussel D, Voituron Y. Alteration of mitochondrial efficiency affects oxidative balance, development and growth in frog (Rana temporaria) tadpoles. ACTA ACUST UNITED AC 2012; 215:863-9. [PMID: 22323209 DOI: 10.1242/jeb.062745] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Mitochondria are known to play a central role in life history processes, being the main source of reactive oxygen species (ROS), which promote oxidative constraint. Surprisingly, although the main role of the mitochondria is to produce ATP, the plasticity of mitochondrial ATP generation has received little attention in life history studies. Yet, mitochondrial energy transduction represents the physiological link between environmental resources and energy allocated to animal performance. Studying both facets of mitochondrial functioning (ATP and ROS production) would allow better understanding of the proximate mechanisms underlying life history. We have experimentally modulated the mitochondrial capacity to generate ROS and ATP during larval development of Rana temporaria tadpoles, via chronic exposure (34 days) to a mitochondrial uncoupler (2,4-dinitrophenol, dNP). The aim was to better understand the impact of mitochondrial uncoupling on both responses in terms of oxidative balance, energy input (oxygen and feeding consumption) and energy output (growth and development of the tadpole). Exposure to 2,4-dNP reduced mitochondrial ROS generation, total antioxidant defences and oxidative damage in treated tadpoles compared with controls. Despite the beneficial effect of dNP on oxidative status, development and growth rates of treated tadpoles were lower than those in the control group. Treatment of tadpoles with 2,4-dNP promoted a mild mitochondrial uncoupling and enhanced metabolic rate. These tadpoles did not increase their food consumption, and thus failed to compensate for the energy loss elicited by the decrease in the efficiency of ATP production. These data suggest that the cost of ATP production, rather than the oxidative balance, is the parameter that constrains growth/development of tadpoles, highlighting the central role of energy transduction in larval performance.
Collapse
Affiliation(s)
- Karine Salin
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés (U.M.R. CNRS 5023), 43 Bvd 11 Novembre 1918, F-69622 Villeurbanne Cedex, France.
| | | | | | | | | |
Collapse
|
34
|
Gómez-Durán A, Pacheu-Grau D, Martínez-Romero I, López-Gallardo E, López-Pérez MJ, Montoya J, Ruiz-Pesini E. Oxidative phosphorylation differences between mitochondrial DNA haplogroups modify the risk of Leber's hereditary optic neuropathy. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1216-22. [PMID: 22561905 DOI: 10.1016/j.bbadis.2012.04.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 03/12/2012] [Accepted: 04/20/2012] [Indexed: 01/12/2023]
Abstract
Leber's hereditary optic neuropathy is a maternally inherited optic atrophy caused by mitochondrial DNA point mutations. Previous epidemiological studies have shown that individuals from mitochondrial genetic backgrounds (haplogroups) J/Uk and H have a higher and a lower risk, respectively, of suffering this disorder. To analyze the bases of these associations at cellular and molecular levels, functional studies with cybrids provide high quality evidence. Cybrids from haplogroup J contain less mitochondrial deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) and synthesize a smaller amount of mitochondrial DNA-encoded polypeptides than those from haplogroup H. Haplogroup J cybrids also display lower oxygen consumption, mitochondrial inner membrane potential and total adenosine-5'-triphosphate (ATP) levels. Moreover, mitochondrial DNA levels correlate with many parameters of the oxidative phosphorylation system. These results suggest that the mitochondrial DNA amount determines oxidative phosphorylation capacity and, along with other recently published observations, support the possibility that mitochondrial DNA levels may be responsible for the bias of the disorder toward males, for the incomplete penetrance of mutations causing Leber's hereditary optic neuropathy and for the association of the disease with particular mitochondrial DNA haplogroups.
Collapse
Affiliation(s)
- Aurora Gómez-Durán
- Departamento de Bioquímica, Biología Molecular y Celular, Centro de Investigaciones Biomédicas En Red de Enfermedades Raras, Universidad de Zaragoza, Spain
| | | | | | | | | | | | | |
Collapse
|
35
|
Jackson EK, Ren J, Cheng D, Mi Z. Extracellular cAMP-adenosine pathways in the mouse kidney. Am J Physiol Renal Physiol 2011; 301:F565-73. [PMID: 21653635 PMCID: PMC3174555 DOI: 10.1152/ajprenal.00094.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 05/31/2011] [Indexed: 01/22/2023] Open
Abstract
The renal extracellular 2',3'-cAMP-adenosine and 3',5'-cAMP-adenosine pathways (extracellular cAMPs→AMPs→adenosine) may contribute to renal adenosine production. Because mouse kidneys provide opportunities to investigate renal adenosine production in genetically modified kidneys, it is important to determine whether mouse kidneys express these cAMP-adenosine pathways. We administered (renal artery) 2',3'-cAMP and 3',5'-cAMP to isolated, perfused mouse kidneys and measured renal venous secretion rates of 2',3'-cAMP, 3',5'-cAMP, 2'-AMP, 3'-AMP, 5'-AMP, adenosine, and inosine. Arterial infusions of 2',3'-cAMP increased (P < 0.0001) the mean venous secretion of 2'-AMP (390-fold), 3'-AMP (497-fold), adenosine (18-fold), and inosine (adenosine metabolite; 7-fold), but they did not alter 5'-AMP secretion. Infusions of 3',5'-cAMP did not affect venous secretion of 2'-AMP or 3'-AMP, but they increased (P < 0.0001) secretion of 5'-AMP (5-fold), adenosine (17-fold), and inosine (6-fold). Energy depletion (metabolic inhibitors) increased the secretion of 2',3'-cAMP (8-fold, P = 0.0081), 2'-AMP (4-fold, P = 0.0028), 3'-AMP (4-fold, P = 0.0270), 5'-AMP (3-fold, P = 0.0662), adenosine (2-fold, P = 0.0317), and inosine (7-fold, P = 0.0071), but it did not increase 3',5'-cAMP secretion. The 2',3'-cAMP-adenosine pathway was quantitatively similar in CD73 -/- vs. +/+ kidneys. However, 3',5'-cAMP induced a 6.7-fold greater increase in 5'-AMP, an attenuated increase (61% reduction) in inosine and a similar increase in adenosine in CD73 -/- vs. CD73 +/+ kidneys. In mouse kidneys, 1) 2',3'-cAMP and 3',5'-cAMP are metabolized to their corresponding AMPs, which are subsequently metabolized to adenosine; 2) energy depletion activates the 2',3'-cAMP-adenosine, but not the 3',5'-cAMP-adenosine, pathway; and 3) although CD73 is involved in the 3',5'-AMP-adenosine pathway, alternative pathways of 5'-AMP metabolism and reduced metabolism of adenosine to inosine compensate for life-long deficiency of CD73.
Collapse
Affiliation(s)
- Edwin K Jackson
- Dept. of Pharmacology and Chemical Biology, Univ. of Pittsburgh School of Medicine, PA 15219, USA.
| | | | | | | |
Collapse
|
36
|
Baffy G, Derdak Z, Robson SC. Mitochondrial recoupling: a novel therapeutic strategy for cancer? Br J Cancer 2011; 105:469-74. [PMID: 21712825 PMCID: PMC3170958 DOI: 10.1038/bjc.2011.245] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent findings link metabolic transformation of cancer cells to aberrant functions of mitochondrial uncoupling proteins (UCPs). By inducing proton leak, UCPs interfere with mitochondrial synthesis of adenosine 5'-triphosphate, which is also a key determinant of glycolytic pathways. In addition, UCP suppress the generation of superoxide, a byproduct of mitochondrial electron transport and a major source of oxidative stress. The near ubiquitous UCP2 becomes highly abundant in some cancers and may advance metabolic reprogramming, further disrupt tumour suppression, and promote chemoresistance. Here we review current evidence to suggest that inhibition of mitochondrial uncoupling may eliminate these responses and reveal novel anti-cancer strategies.
Collapse
Affiliation(s)
- G Baffy
- Department of Medicine, VA Boston Healthcare System and Brigham and Women's Hospital, Harvard Medical School, 150 S Huntington Avenue, Room A6-46, Boston, MA 02130, USA.
| | | | | |
Collapse
|
37
|
Tight control of mitochondrial membrane potential by cytochrome c oxidase. Mitochondrion 2010; 11:334-41. [PMID: 21147274 DOI: 10.1016/j.mito.2010.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 11/24/2010] [Accepted: 12/03/2010] [Indexed: 11/23/2022]
Abstract
In the present work we have critically examined the use of the KCN-titration technique in the study of the control of the cellular respiration by cytochrome c oxidase (COX) in the presence of the mitochondrial membrane potential (Δψ(mito)) in HepG2 cells. We clearly show that the apparent high inhibition threshold of COX in the presence of maximal Δψ(mito) is due to the KCN-induced decrease of Δψ(mito) and not to a low control of COX on the mitochondrial respiration. The tight control exerted by COX on the Δψ(mito) provides further insights for understanding the pathogenetic mechanisms associated with mitochondrial defects in human neuromuscular degenerative disorders.
Collapse
|
38
|
Chevrollier A, Loiseau D, Reynier P, Stepien G. Adenine nucleotide translocase 2 is a key mitochondrial protein in cancer metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1807:562-7. [PMID: 20950584 DOI: 10.1016/j.bbabio.2010.10.008] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 10/05/2010] [Accepted: 10/05/2010] [Indexed: 12/12/2022]
Abstract
Adenine nucleotide translocase (ANT), a mitochondrial protein that facilitates the exchange of ADP and ATP across the mitochondrial inner membrane, plays an essential role in cellular energy metabolism. Human ANT presents four isoforms (ANT1-4), each with a specific expression depending on the nature of the tissue, cell type, developmental stage and status of cell proliferation. Thus, ANT1 is specific to muscle and brain tissues; ANT2 occurs mainly in proliferative, undifferentiated cells; ANT3 is ubiquitous; and ANT4 is found in germ cells. ANT1 and ANT3 export the ATP produced by oxidative phosphorylation (OxPhos) from the mitochondria into the cytosol while importing ADP. In contrast, the expression of ANT2, which is linked to the rate of glycolytic metabolism, is an important indicator of carcinogenesis. In fact, cancers are characterized by major metabolic changes that switch cells from the normally dual oxidative and glycolytic metabolisms to an almost exclusively glycolytic metabolism. When OxPhos activity is impaired, ANT2 imports glycolytically produced ATP into the mitochondria. In the mitochondrial matrix, the F1F0-ATPase complex hydrolyzes the ATP, pumping out a proton into the intermembrane space. The reverse operations of ANT2 and F1F0-ATPase under glycolytic conditions contribute to maintaining the mitochondrial membrane potential, ensuring cell survival and proliferation. Unlike the ANT1 and ANT3 isoforms, ANT2 is not pro-apoptotic and may therefore contribute to carcinogenesis. Since the expression of ANT2 is closely linked to the mitochondrial bioenergetics of tumors, it should be taken into account for individualizing cancer treatments and for the development of anticancer strategies.
Collapse
|
39
|
Gómez-Durán A, Pacheu-Grau D, López-Gallardo E, Díez-Sánchez C, Montoya J, López-Pérez MJ, Ruiz-Pesini E. Unmasking the causes of multifactorial disorders: OXPHOS differences between mitochondrial haplogroups. Hum Mol Genet 2010; 19:3343-53. [PMID: 20566709 DOI: 10.1093/hmg/ddq246] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many epidemiologic studies have associated human mitochondrial haplogroups to rare mitochondrial diseases like Leber's hereditary optic neuropathy or to more common age-linked disorders such as Parkinson's disease. However, cellular, biochemical and molecular-genetic evidence that is able to explain these associations is very scarce. The etiology of multifactorial diseases is very difficult to sort out because such diseases are due to a combination of genetic and environmental factors that individually only contribute in small part to the development of the illness. Thus, the haplogroup-defining mutations might behave as susceptibility factors, but they could have only a small effect on oxidative phosphorylation (OXPHOS) function. Moreover, these effects would be highly dependent on the 'context' in which the genetic variant is acting. To homogenize this 'context' for mitochondrial DNA (mtDNA) mutations, a cellular approach is available that involves the use of what is known as 'cybrids'. By using this model, we demonstrate that mtDNA and mtRNA levels, mitochondrial protein synthesis, cytochrome oxidase activity and amount, normalized oxygen consumption, mitochondrial inner membrane potential and growth capacity are different in cybrids from the haplogroup H when compared with those of the haplogroup Uk. Thus, these inherited basal differences in OXPHOS capacity can help to explain why some individuals more quickly reach the bioenergetic threshold below which tissue symptoms appear and progress toward multifactorial disorders. Hence, some population genetic variants in mtDNA contribute to the genetic component of complex disorders. The existence of mtDNA-based OXPHOS differences opens possibilities for the existence of a new field, mitochondrial pharmacogenomics. New sequence accession nos: HM103354-HM103363.
Collapse
Affiliation(s)
- Aurora Gómez-Durán
- Departamento de Bioquímica, Biología Molecular y Celular, Centro de INvestigaciones Biomédicas en Red de Enfermedades Raras, Instituto Aragonés de Ciencias de la Salud, Universidad de Zaragoza, Zaragoza, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Dikov D, Aulbach A, Muster B, Dröse S, Jendrach M, Bereiter-Hahn J. Do UCP2 and mild uncoupling improve longevity? Exp Gerontol 2010; 45:586-95. [PMID: 20332018 DOI: 10.1016/j.exger.2010.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/04/2010] [Accepted: 03/16/2010] [Indexed: 11/18/2022]
Abstract
Mild uncoupling of mitochondrial respiration is considered to prolong life span of organisms by reducing the production of reactive oxygen species (ROS). Experimental evidence against this hypothesis has been brought forward by premature senescence in cell cultures treated with uncouplers. Exposing HUVEC to a mixture of nutritionally important fatty acids (oil extract of chicken yolk) mild uncoupling with "naturally acting substances" was performed. This treatment also resulted in premature senescence although ROS production did not increase. Fatty acids activate uncoupling proteins (UCP) in the inner mitochondrial membrane. UCP2 expression proved to be sensitive to the presence of fatty acids but remains unchanged during the ageing process. UCP3 expression in senescent HUVEC and avUCP expression in senescent CEF were considerably less than in young cultures. No indication for protonophoric reduction of mitochondrial membrane potential was found in UCP2 overexpressing HeLa cells and only little in HUVEC. ROS levels increased instead of being reduced in these cells. Stable transfection with UCP2-GFP was possible only in chick embryo fibroblasts and HeLa cells and resulted in decreased proliferation. Stable transfection of HUVEC with UCP2-GFP resulted in death of cultures within one or two weeks. The reason for this behaviour most probably is apoptosis preceded by mitochondrial fragmentation and loss of membrane potential.
Collapse
Affiliation(s)
- Daniel Dikov
- Institute for Cell Biology and Neurosciences, Biocenter. Goethe University, Max von Lauestrasse 9, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Jahnke VE, Sabido O, Defour A, Castells J, Lefai E, Roussel D, Freyssenet D. Evidence for mitochondrial respiratory deficiency in rat rhabdomyosarcoma cells. PLoS One 2010; 5:e8637. [PMID: 20072609 PMCID: PMC2797644 DOI: 10.1371/journal.pone.0008637] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 12/11/2009] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Mitochondria can sense signals linked to variations in energy demand to regulate nuclear gene expression. This retrograde signaling pathway is presumed to be involved in the regulation of myoblast proliferation and differentiation. Rhabdomyosarcoma cells are characterized by their failure to both irreversibly exit the cell cycle and complete myogenic differentiation. However, it is currently unknown whether mitochondria are involved in the failure of rhabdomyosarcoma cells to differentiate. METHODOLOGY/PRINCIPAL FINDINGS Mitochondrial biogenesis and metabolism were studied in rat L6E9 myoblasts and R1H rhabdomyosacoma cells during the cell cycle and after 36 hours of differentiation. Using a combination of flow cytometry, polarographic and molecular analyses, we evidenced a marked decrease in the cardiolipin content of R1H cells cultured in growth and differentiation media, together with a significant increase in the content of mitochondrial biogenesis factors and mitochondrial respiratory chain proteins. Altogether, these data indicate that the mitochondrial inner membrane composition and the overall process of mitochondrial biogenesis are markedly altered in R1H cells. Importantly, the dysregulation of protein-to-cardiolipin ratio was associated with major deficiencies in both basal and maximal mitochondrial respiration rates. This deficiency in mitochondrial respiration probably contributes to the inability of R1H cells to decrease mitochondrial H2O2 level at the onset of differentiation. CONCLUSION/SIGNIFICANCE A defect in the regulation of mitochondrial biogenesis and mitochondrial metabolism may thus be an epigenetic mechanism that may contribute to the tumoral behavior of R1H cells. Our data underline the importance of mitochondria in the regulation of myogenic differentiation.
Collapse
Affiliation(s)
- Vanessa E. Jahnke
- Université de Lyon, Université Jean Monnet, Laboratoire de Physiologie de l'Exercice EA4338, Saint Etienne, France
| | - Odile Sabido
- Université de Lyon, Université Jean Monnet, Laboratoire de Physiologie de l'Exercice EA4338, Saint Etienne, France
- Université de Lyon, Université Jean Monnet, Centre Commun de Cytométrie en Flux, Saint Etienne, France
| | - Aurélia Defour
- Université de Lyon, Université Jean Monnet, Laboratoire de Physiologie de l'Exercice EA4338, Saint Etienne, France
| | - Josiane Castells
- Université de Lyon, Université Jean Monnet, Laboratoire de Physiologie de l'Exercice EA4338, Saint Etienne, France
| | - Etienne Lefai
- Université de Lyon, Université Claude Bernard Lyon 1, Régulations Métaboliques Nutrition et Diabètes INSERM U870, Oullins, France
| | - Damien Roussel
- Université de Lyon, Université Claude Bernard Lyon 1, Laboratoire de Physiologie Intégrative Cellulaire et Moléculaire CNRS U5123, Villeurbanne, France
| | - Damien Freyssenet
- Université de Lyon, Université Jean Monnet, Laboratoire de Physiologie de l'Exercice EA4338, Saint Etienne, France
| |
Collapse
|
42
|
Chan SL, Wei Z, Chigurupati S, Tu W. Compromised respiratory adaptation and thermoregulation in aging and age-related diseases. Ageing Res Rev 2010; 9:20-40. [PMID: 19800420 DOI: 10.1016/j.arr.2009.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 09/22/2009] [Accepted: 09/23/2009] [Indexed: 02/04/2023]
Abstract
Mitochondrial dysfunction and reactive oxygen species (ROS) production are at the heart of the aging process and are thought to underpin age-related diseases. Mitochondria are not only the primary energy-generating system but also the dominant cellular source of metabolically derived ROS. Recent studies unravel the existence of mechanisms that serve to modulate the balance between energy metabolism and ROS production. Among these is the regulation of proton conductance across the inner mitochondrial membrane that affects the efficiency of respiration and heat production. The field of mitochondrial respiration research has provided important insight into the role of altered energy balance in obesity and diabetes. The notion that respiration and oxidative capacity are mechanistically linked is making significant headway into the field of aging and age-related diseases. Here we review the regulation of cellular energy and ROS balance in biological systems and survey some of the recent relevant studies that suggest that respiratory adaptation and thermodynamics are important in aging and age-related diseases.
Collapse
|
43
|
Loiseau D, Morvan D, Chevrollier A, Demidem A, Douay O, Reynier P, Stepien G. Mitochondrial bioenergetic background confers a survival advantage to HepG2 cells in response to chemotherapy. Mol Carcinog 2009; 48:733-41. [PMID: 19347860 DOI: 10.1002/mc.20539] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cancer cells mainly rely on glycolysis for energetic needs, and mitochondrial ATP production is almost inactive. However, cancer cells require the integrity of mitochondrial functions for their survival, such as the maintenance of the internal membrane potential gradient (DeltaPsim). It thus may be predicted that DeltaPsim regeneration should depend on cellular capability to produce sufficient ATP by upregulating glycolysis or recruiting oxidative phosphorylation (OXPHOS). To investigate this hypothesis, we compared the response to an anticancer agent chloroethylnitrosourea (CENU) of two transformed cell lines: HepG2 (hepatocarcinoma) with a partially differentiated phenotype and 143B (osteosarcoma) with an undifferentiated one. These cells types differ by their mitochondrial OXPHOS background; the most severely impaired being that of 143B cells. Treatment effects were tested on cell proliferation, O(2) consumption/ATP production coupling, DeltaPsim maintenance, and global metabolite profiling by NMR spectroscopy. Our results showed an OXPHOS uncoupling and a lowered DeltaPsim, leading to an increased energy request to regenerate DeltaPsim in both models. However, energy request could not be met by undifferentiated cells 143B, which ATP content decreased after 48 h leading to cell death, while partially differentiated cells (HepG2) could activate their oxidative metabolism and escape chemotherapy. We propose that mitochondrial OXPHOS background confers a survival advantage to more differentiated cells in response to chemotherapy. This suggests that the mitochondrial bioenergetic background of tumors should be considered for anticancer treatment personalization.
Collapse
|
44
|
Guillet V, Gueguen N, Verny C, Ferre M, Homedan C, Loiseau D, Procaccio V, Amati-Bonneau P, Bonneau D, Reynier P, Chevrollier A. Adenine nucleotide translocase is involved in a mitochondrial coupling defect in MFN2-related Charcot-Marie-Tooth type 2A disease. Neurogenetics 2009; 11:127-33. [PMID: 19618221 DOI: 10.1007/s10048-009-0207-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 06/29/2009] [Indexed: 11/26/2022]
Abstract
Charcot-Marie-Tooth type 2A disease (CMT2A), a dominantly inherited peripheral neuropathy, is caused by mutations in MFN2, a mitochondrial fusion protein. Having previously demonstrated a mitochondrial coupling defect in CMT2A patients' fibroblasts, we here investigate mitochondrial oxygen consumption and the expression of adenine nucleotide translocase (ANT) and uncoupling proteins from eight other patients with the disease. The mitochondrial uncoupling was associated with a higher respiratory rate, essentially involving complex II proteins. Furthermore, a twofold increase in the expression of ANT led to the reduced efficiency of oxidative phosphorylation in CMT2A cells, suggesting that MFN2 plays a role in controlling ATP/ADP exchanges.
Collapse
|
45
|
Dumas JF, Simard G, Flamment M, Ducluzeau PH, Ritz P. Is skeletal muscle mitochondrial dysfunction a cause or an indirect consequence of insulin resistance in humans? DIABETES & METABOLISM 2009; 35:159-67. [DOI: 10.1016/j.diabet.2009.02.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 02/21/2009] [Accepted: 02/23/2009] [Indexed: 12/25/2022]
|
46
|
Bernatoniene J, Trumbeckaite S, Majiene D, Baniene R, Baliutyte G, Savickas A, Toleikis A. The effect of crataegus fruit extract and some of its flavonoids on mitochondrial oxidative phosphorylation in the heart. Phytother Res 2009; 23:1701-7. [DOI: 10.1002/ptr.2815] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Samudio I, Fiegl M, McQueen T, Clise-Dwyer K, Andreeff M. The warburg effect in leukemia-stroma cocultures is mediated by mitochondrial uncoupling associated with uncoupling protein 2 activation. Cancer Res 2008; 68:5198-205. [PMID: 18593920 DOI: 10.1158/0008-5472.can-08-0555] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In 1956, Otto Warburg proposed that the origin of cancer cells was closely linked to a permanent respiratory defect that bypassed the Pasteur effect (i.e., the inhibition of anaerobic fermentation by oxygen). Since then, permanent defects in oxygen consumption that could explain the dependence of cancer cells on aerobic glycolysis have not been identified. Here, we show that under normoxic conditions exposure of leukemia cells to bone marrow-derived mesenchymal stromal cells (MSC) promotes accumulation of lactate in the culture medium and reduces mitochondrial membrane potential (DeltaPsiM) in both cell types. Notably, the consumption of glucose was not altered in cocultures, suggesting that the accumulation of lactate was the result of reduced pyruvate metabolism. Interestingly, the decrease in DeltaPsiM was mediated by mitochondrial uncoupling in leukemia cells and was accompanied by increased expression of uncoupling protein 2 (UCP2). HL60 cells fail to increase UCP2 expression, are not uncoupled after coculture, and do not exhibit increased aerobic glycolysis, whereas small interfering RNA-mediated suppression of UCP2 in OCI-AML3 cells reversed mitochondrial uncoupling and aerobic glycolysis elicited by MSC. Taken together, these data suggest that microenvironment activation of highly conserved mammalian UCPs may facilitate the Warburg effect in the absence of permanent respiratory impairment.
Collapse
Affiliation(s)
- Ismael Samudio
- Department of Stem Cell Transplantation and Cellular Therapy, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
48
|
Benard G, Rossignol R. Ultrastructure of the mitochondrion and its bearing on function and bioenergetics. Antioxid Redox Signal 2008; 10:1313-42. [PMID: 18435594 DOI: 10.1089/ars.2007.2000] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The recently ascertained network and dynamic organization of the mitochondrion, as well as the demonstration of energy proteins and metabolites subcompartmentalization, have led to a reconsideration of the relationships between organellar form and function. In particular, the impact of mitochondrial morphological changes on bioenergetics is inseparable. Several observations indicate that mitochondrial energy production may be controlled by structural rearrangements of the organelle both interiorly and globally, including the remodeling of cristae morphology and elongation or fragmentation of the tubular network organization, respectively. These changes are mediated by fusion or fission reactions in response to physiological signals that remain unidentified. They lead to important changes in the internal diffusion of energy metabolites, the sequestration and conduction of the electric membrane potential (Delta Psi), and possibly the delivery of newly synthesized ATP to various cellular areas. Moreover, the physiological or even pathological context also determines the morphology of the mitochondrion, suggesting a tight and mutual control between mitochondrial form and bioenergetics. In this review, we delve into the link between mitochondrial structure and energy metabolism.
Collapse
|
49
|
Plecitá-Hlavatá L, Lessard M, Šantorová J, Bewersdorf J, Ježek P. Mitochondrial oxidative phosphorylation and energetic status are reflected by morphology of mitochondrial network in INS-1E and HEP-G2 cells viewed by 4Pi microscopy. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2008; 1777:834-46. [DOI: 10.1016/j.bbabio.2008.04.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 03/31/2008] [Accepted: 04/01/2008] [Indexed: 10/22/2022]
|
50
|
Pearen MA, Myers SA, Raichur S, Ryall JG, Lynch GS, Muscat GEO. The orphan nuclear receptor, NOR-1, a target of beta-adrenergic signaling, regulates gene expression that controls oxidative metabolism in skeletal muscle. Endocrinology 2008; 149:2853-65. [PMID: 18325999 DOI: 10.1210/en.2007-1202] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
beta 1-3-Adrenoreceptor (AR)-deficient mice are unable to regulate energy expenditure and develop diet-induced obesity on a high-fat diet. We determined previously that beta2-AR agonist treatment activated expression of the mRNA encoding the orphan nuclear receptor, NOR-1, in muscle cells and plantaris muscle. Here we show that beta2-AR agonist treatment significantly and transiently activated the expression of NOR-1 (and the other members of the NR4A subgroup) in slow-twitch oxidative soleus muscle and fast-twitch glycolytic tibialis anterior muscle. The activation induced by beta-adrenergic signaling is consistent with the involvement of protein kinase A, MAPK, and phosphorylation of cAMP response element-binding protein. Stable cell lines transfected with a silent interfering RNA targeting NOR-1 displayed decreased palmitate oxidation and lactate accumulation. In concordance with these observations, ATP production in the NOR-1 silent interfering RNA (but not control)-transfected cells was resistant to (azide-mediated) inhibition of oxidative metabolism and expressed significantly higher levels of hypoxia inducible factor-1alpha. In addition, we observed the repression of genes that promote fatty acid oxidation (peroxisomal proliferator-activated receptor-gamma coactivator-1alpha/beta and lipin-1alpha) and trichloroacetic acid cycle-mediated carbohydrate (pyruvate) oxidation [pyruvate dehydrogenase phosphatase 1 regulatory and catalytic subunits (pyruvate dehydrogenase phosphatases-1r and -c)]. Furthermore, we observed that beta2-AR agonist administration in mouse skeletal muscle induced the expression of genes that activate fatty acid oxidation and modulate pyruvate use, including PGC-1alpha, lipin-1alpha, FOXO1, and PDK4. Finally, we demonstrate that NOR-1 is recruited to the lipin-1alpha and PDK-4 promoters, and this is consistent with NOR-1-mediated regulation of these genes. In conclusion, NOR-1 is necessary for oxidative metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Michael A Pearen
- Institute for Molecular Bioscience, The University of Queensland, Queensland 4072, Australia
| | | | | | | | | | | |
Collapse
|