Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.381
Filter
Add more filters

Publication year range
1.
Mol Cell ; 84(5): 981-989.e7, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38295803

ABSTRACT

Coenzyme Q (CoQ) is a redox lipid that fulfills critical functions in cellular bioenergetics and homeostasis. CoQ is synthesized by a multi-step pathway that involves several COQ proteins. Two steps of the eukaryotic pathway, the decarboxylation and hydroxylation of position C1, have remained uncharacterized. Here, we provide evidence that these two reactions occur in a single oxidative decarboxylation step catalyzed by COQ4. We demonstrate that COQ4 complements an Escherichia coli strain deficient for C1 decarboxylation and hydroxylation and that COQ4 displays oxidative decarboxylation activity in the non-CoQ producer Corynebacterium glutamicum. Overall, our results substantiate that COQ4 contributes to CoQ biosynthesis, not only via its previously proposed structural role but also via the oxidative decarboxylation of CoQ precursors. These findings fill a major gap in the knowledge of eukaryotic CoQ biosynthesis and shed light on the pathophysiology of human primary CoQ deficiency due to COQ4 mutations.


Subject(s)
Eukaryotic Cells , Ubiquinone , Humans , Decarboxylation , Eukaryotic Cells/metabolism , Oxidation-Reduction , Escherichia coli/genetics , Escherichia coli/metabolism , Oxidative Stress , Mitochondrial Proteins/metabolism
2.
Physiol Rev ; 104(4): 1533-1610, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38722242

ABSTRACT

Coenzyme Q (CoQ), also known as ubiquinone, comprises a benzoquinone head group and a long isoprenoid side chain. It is thus extremely hydrophobic and resides in membranes. It is best known for its complex function as an electron transporter in the mitochondrial electron transport chain (ETC) but is also required for several other crucial cellular processes. In fact, CoQ appears to be central to the entire redox balance of the cell. Remarkably, its structure and therefore its properties have not changed from bacteria to vertebrates. In metazoans, it is synthesized in all cells and is found in most, and maybe all, biological membranes. CoQ is also known as a nutritional supplement, mostly because of its involvement with antioxidant defenses. However, whether there is any health benefit from oral consumption of CoQ is not well established. Here we review the function of CoQ as a redox-active molecule in the ETC and other enzymatic systems, its role as a prooxidant in reactive oxygen species generation, and its separate involvement in antioxidant mechanisms. We also review CoQ biosynthesis, which is particularly complex because of its extreme hydrophobicity, as well as the biological consequences of primary and secondary CoQ deficiency, including in human patients. Primary CoQ deficiency is a rare inborn condition due to mutation in CoQ biosynthetic genes. Secondary CoQ deficiency is much more common, as it accompanies a variety of pathological conditions, including mitochondrial disorders as well as aging. In this context, we discuss the importance, but also the great difficulty, of alleviating CoQ deficiency by CoQ supplementation.


Subject(s)
Mitochondria , Ubiquinone , Ubiquinone/metabolism , Ubiquinone/analogs & derivatives , Humans , Animals , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Oxidation-Reduction , Antioxidants/metabolism , Muscle Weakness/metabolism , Reactive Oxygen Species/metabolism , Ataxia/metabolism
3.
Nature ; 631(8019): 232-239, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38811722

ABSTRACT

Mitochondria play a pivotal part in ATP energy production through oxidative phosphorylation, which occurs within the inner membrane through a series of respiratory complexes1-4. Despite extensive in vitro structural studies, determining the atomic details of their molecular mechanisms in physiological states remains a major challenge, primarily because of loss of the native environment during purification. Here we directly image porcine mitochondria using an in situ cryo-electron microscopy approach. This enables us to determine the structures of various high-order assemblies of respiratory supercomplexes in their native states. We identify four main supercomplex organizations: I1III2IV1, I1III2IV2, I2III2IV2 and I2III4IV2, which potentially expand into higher-order arrays on the inner membranes. These diverse supercomplexes are largely formed by 'protein-lipids-protein' interactions, which in turn have a substantial impact on the local geometry of the surrounding membranes. Our in situ structures also capture numerous reactive intermediates within these respiratory supercomplexes, shedding light on the dynamic processes of the ubiquinone/ubiquinol exchange mechanism in complex I and the Q-cycle in complex III. Structural comparison of supercomplexes from mitochondria treated under different conditions indicates a possible correlation between conformational states of complexes I and III, probably in response to environmental changes. By preserving the native membrane environment, our approach enables structural studies of mitochondrial respiratory supercomplexes in reaction at high resolution across multiple scales, from atomic-level details to the broader subcellular context.


Subject(s)
Cell Respiration , Electron Transport Complex III , Electron Transport Complex I , Mitochondria , Animals , Cryoelectron Microscopy , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Electron Transport Complex I/ultrastructure , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Electron Transport Complex III/ultrastructure , Mitochondria/metabolism , Mitochondria/chemistry , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/ultrastructure , Models, Molecular , Oxidative Phosphorylation , Swine , Ubiquinone/analogs & derivatives , Ubiquinone/chemistry , Ubiquinone/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism
4.
Mol Cell ; 82(22): 4307-4323.e10, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36306796

ABSTRACT

Coenzyme Q (CoQ) is a redox-active lipid essential for core metabolic pathways and antioxidant defense. CoQ is synthesized upon the mitochondrial inner membrane by an ill-defined "complex Q" metabolon. Here, we present structure-function analyses of a lipid-, substrate-, and NADH-bound complex comprising two complex Q subunits: the hydroxylase COQ7 and the lipid-binding protein COQ9. We reveal that COQ7 adopts a ferritin-like fold with a hydrophobic channel whose substrate-binding capacity is enhanced by COQ9. Using molecular dynamics, we further show that two COQ7:COQ9 heterodimers form a curved tetramer that deforms the membrane, potentially opening a pathway for the CoQ intermediates to translocate from the bilayer to the proteins' lipid-binding sites. Two such tetramers assemble into a soluble octamer with a pseudo-bilayer of lipids captured within. Together, these observations indicate that COQ7 and COQ9 cooperate to access hydrophobic precursors within the membrane and coordinate subsequent synthesis steps toward producing CoQ.


Subject(s)
Mitochondrial Membranes , Ubiquinone , Humans , Ubiquinone/chemistry , Mitochondrial Membranes/metabolism , Carrier Proteins , Lipids
5.
Nature ; 618(7964): 358-364, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225987

ABSTRACT

The ability to switch between different lifestyles allows bacterial pathogens to thrive in diverse ecological niches1,2. However, a molecular understanding of their lifestyle changes within the human host is lacking. Here, by directly examining bacterial gene expression in human-derived samples, we discover a gene that orchestrates the transition between chronic and acute infection in the opportunistic pathogen Pseudomonas aeruginosa. The expression level of this gene, here named sicX, is the highest of the P. aeruginosa genes expressed in human chronic wound and cystic fibrosis infections, but it is expressed at extremely low levels during standard laboratory growth. We show that sicX encodes a small RNA that is strongly induced by low-oxygen conditions and post-transcriptionally regulates anaerobic ubiquinone biosynthesis. Deletion of sicX causes P. aeruginosa to switch from a chronic to an acute lifestyle in multiple mammalian models of infection. Notably, sicX is also a biomarker for this chronic-to-acute transition, as it is the most downregulated gene when a chronic infection is dispersed to cause acute septicaemia. This work solves a decades-old question regarding the molecular basis underlying the chronic-to-acute switch in P. aeruginosa and suggests oxygen as a primary environmental driver of acute lethality.


Subject(s)
Acute Disease , Chronic Disease , Genes, Bacterial , Oxygen , Pseudomonas Infections , Pseudomonas aeruginosa , RNA, Bacterial , Animals , Humans , Oxygen/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Cystic Fibrosis/microbiology , Wounds and Injuries/microbiology , Ubiquinone/biosynthesis , Anaerobiosis , Genes, Bacterial/genetics , Sepsis/complications , Sepsis/microbiology
6.
Nature ; 619(7969): 371-377, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380771

ABSTRACT

Ferroptosis is evolving as a highly promising approach to combat difficult-to-treat tumour entities including therapy-refractory and dedifferentiating cancers1-3. Recently, ferroptosis suppressor protein-1 (FSP1), along with extramitochondrial ubiquinone or exogenous vitamin K and NAD(P)H/H+ as an electron donor, has been identified as the second ferroptosis-suppressing system, which efficiently prevents lipid peroxidation independently of the cyst(e)ine-glutathione (GSH)-glutathione peroxidase 4 (GPX4) axis4-6. To develop FSP1 inhibitors as next-generation therapeutic ferroptosis inducers, here we performed a small molecule library screen and identified the compound class of 3-phenylquinazolinones (represented by icFSP1) as potent FSP1 inhibitors. We show that icFSP1, unlike iFSP1, the first described on-target FSP1 inhibitor5, does not competitively inhibit FSP1 enzyme activity, but instead triggers subcellular relocalization of FSP1 from the membrane and FSP1 condensation before ferroptosis induction, in synergism with GPX4 inhibition. icFSP1-induced FSP1 condensates show droplet-like properties consistent with phase separation, an emerging and widespread mechanism to modulate biological activity7. N-terminal myristoylation, distinct amino acid residues and intrinsically disordered, low-complexity regions in FSP1 were identified to be essential for FSP1-dependent phase separation in cells and in vitro. We further demonstrate that icFSP1 impairs tumour growth and induces FSP1 condensates in tumours in vivo. Hence, our results suggest that icFSP1 exhibits a unique mechanism of action and synergizes with ferroptosis-inducing agents to potentiate the ferroptotic cell death response, thus providing a rationale for targeting FSP1-dependent phase separation as an efficient anti-cancer therapy.


Subject(s)
Apoptosis Regulatory Proteins , Ferroptosis , Mitochondrial Proteins , Humans , Amino Acids/metabolism , Cysteine/metabolism , Ferroptosis/drug effects , Glutathione/metabolism , NAD/metabolism , NADP/metabolism , Neoplasms/drug therapy , Quinazolines/pharmacology , Small Molecule Libraries , Ubiquinone/metabolism , Vitamin K/metabolism , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism
7.
Nature ; 621(7977): 179-187, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648857

ABSTRACT

Tissue resident memory CD8+ T (TRM) cells offer rapid and long-term protection at sites of reinfection1. Tumour-infiltrating lymphocytes with characteristics of TRM cells maintain enhanced effector functions, predict responses to immunotherapy and accompany better prognoses2,3. Thus, an improved understanding of the metabolic strategies that enable tissue residency by T cells could inform new approaches to empower immune responses in tissues and solid tumours. Here, to systematically define the basis for the metabolic reprogramming supporting TRM cell differentiation, survival and function, we leveraged in vivo functional genomics, untargeted metabolomics and transcriptomics of virus-specific memory CD8+ T cell populations. We found that memory CD8+ T cells deployed a range of adaptations to tissue residency, including reliance on non-steroidal products of the mevalonate-cholesterol pathway, such as coenzyme Q, driven by increased activity of the transcription factor SREBP2. This metabolic adaptation was most pronounced in the small intestine, where TRM cells interface with dietary cholesterol and maintain a heightened state of activation4, and was shared by functional tumour-infiltrating lymphocytes in diverse tumour types in mice and humans. Enforcing synthesis of coenzyme Q through deletion of Fdft1 or overexpression of PDSS2 promoted mitochondrial respiration, memory T cell formation following viral infection and enhanced antitumour immunity. In sum, through a systematic exploration of TRM cell metabolism, we reveal how these programs can be leveraged to fuel memory CD8+ T cell formation in the context of acute infections and enhance antitumour immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Neoplasms , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Respiration , Cholesterol/metabolism , Cholesterol/pharmacology , Immunologic Memory , Intestine, Small/drug effects , Intestine, Small/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Metabolomics , Mevalonic Acid/metabolism , Neoplasms/immunology , Ubiquinone/metabolism , Virus Diseases/immunology , Viruses/immunology , Mitochondria/metabolism
8.
Annu Rev Biochem ; 82: 551-75, 2013.
Article in English | MEDLINE | ID: mdl-23527692

ABSTRACT

Complex I (NADH:ubiquinone oxidoreductase) is crucial for respiration in many aerobic organisms. In mitochondria, it oxidizes NADH from the tricarboxylic acid cycle and ß-oxidation, reduces ubiquinone, and transports protons across the inner membrane, contributing to the proton-motive force. It is also a major contributor to cellular production of reactive oxygen species. The redox reaction of complex I is catalyzed in the hydrophilic domain; it comprises NADH oxidation by a flavin mononucleotide, intramolecular electron transfer along a chain of iron-sulfur clusters, and ubiquinone reduction. Redox-coupled proton translocation in the membrane domain requires long-range energy transfer through the protein complex, and the molecular mechanisms that couple the redox and proton-transfer half-reactions are currently unknown. This review evaluates extant data on the mechanisms of energy transduction and superoxide production by complex I, discusses contemporary mechanistic models, and explores how mechanistic studies may contribute to understanding the roles of complex I dysfunctions in human diseases.


Subject(s)
Electron Transport Complex I/metabolism , Flavins/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Ubiquinone/metabolism , Electron Transport , Electron Transport Complex I/chemistry , Humans , Oxidation-Reduction
9.
EMBO J ; 43(2): 168-195, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38212382

ABSTRACT

Coenzyme Q (CoQ) is essential for mitochondrial respiration and required for thermogenic activity in brown adipose tissues (BAT). CoQ deficiency leads to a wide range of pathological manifestations, but mechanistic consequences of CoQ deficiency in specific tissues, such as BAT, remain poorly understood. Here, we show that pharmacological or genetic CoQ deficiency in BAT leads to stress signals causing accumulation of cytosolic mitochondrial RNAs and activation of the eIF2α kinase PKR, resulting in activation of the integrated stress response (ISR) with suppression of UCP1 but induction of FGF21 expression. Strikingly, despite diminished UCP1 levels, BAT CoQ deficiency displays increased whole-body metabolic rates at room temperature and thermoneutrality resulting in decreased weight gain on high-fat diets (HFD). In line with enhanced metabolic rates, BAT and inguinal white adipose tissue (iWAT) interorgan crosstalk caused increased browning of iWAT in BAT-specific CoQ deficient animals. This mitohormesis-like effect depends on the ATF4-FGF21 axis and BAT-secreted FGF21, revealing an unexpected role for CoQ in the modulation of whole-body energy expenditure with wide-ranging implications for primary and secondary CoQ deficiencies.


Subject(s)
Adipose Tissue, Brown , Ataxia , Fibroblast Growth Factors , Mitochondrial Diseases , Muscle Weakness , Animals , Mice , Adipose Tissue, Brown/metabolism , Ubiquinone/metabolism , Ubiquinone/pharmacology , Mitochondrial Diseases/metabolism , Thermogenesis/genetics , Mice, Inbred C57BL
11.
Cell ; 152(3): 504-18, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23374346

ABSTRACT

Protection against oxidative damage caused by excessive reactive oxygen species (ROS) by an antioxidant network is essential for the health of tissues, especially in the cardiovascular system. Here, we identified a gene with important antioxidant features by analyzing a null allele of zebrafish ubiad1, called barolo (bar). bar mutants show specific cardiovascular failure due to oxidative stress and ROS-mediated cellular damage. Human UBIAD1 is a nonmitochondrial prenyltransferase that synthesizes CoQ10 in the Golgi membrane compartment. Loss of UBIAD1 reduces the cytosolic pool of the antioxidant CoQ10 and leads to ROS-mediated lipid peroxidation in vascular cells. Surprisingly, inhibition of eNOS prevents Ubiad1-dependent cardiovascular oxidative damage, suggesting a crucial role for this enzyme and nonmitochondrial CoQ10 in NO signaling. These findings identify UBIAD1 as a nonmitochondrial CoQ10-forming enzyme with specific cardiovascular protective function via the modulation of eNOS activity.


Subject(s)
Dimethylallyltranstransferase/metabolism , Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/metabolism , Ubiquinone/analogs & derivatives , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Dimethylallyltranstransferase/genetics , Golgi Apparatus/metabolism , Heart/embryology , Humans , Myocardium/cytology , Reactive Oxygen Species/metabolism , Ubiquinone/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics
12.
Genes Dev ; 34(7-8): 526-543, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32079652

ABSTRACT

MDM2 and MDMX, negative regulators of the tumor suppressor p53, can work separately and as a heteromeric complex to restrain p53's functions. MDM2 also has pro-oncogenic roles in cells, tissues, and animals that are independent of p53. There is less information available about p53-independent roles of MDMX or the MDM2-MDMX complex. We found that MDM2 and MDMX facilitate ferroptosis in cells with or without p53. Using small molecules, RNA interference reagents, and mutant forms of MDMX, we found that MDM2 and MDMX, likely working in part as a complex, normally facilitate ferroptotic death. We observed that MDM2 and MDMX alter the lipid profile of cells to favor ferroptosis. Inhibition of MDM2 or MDMX leads to increased levels of FSP1 protein and a consequent increase in the levels of coenzyme Q10, an endogenous lipophilic antioxidant. This suggests that MDM2 and MDMX normally prevent cells from mounting an adequate defense against lipid peroxidation and thereby promote ferroptosis. Moreover, we found that PPARα activity is essential for MDM2 and MDMX to promote ferroptosis, suggesting that the MDM2-MDMX complex regulates lipids through altering PPARα activity. These findings reveal the complexity of cellular responses to MDM2 and MDMX and suggest that MDM2-MDMX inhibition might be useful for preventing degenerative diseases involving ferroptosis. Furthermore, they suggest that MDM2/MDMX amplification may predict sensitivity of some cancers to ferroptosis inducers.


Subject(s)
Cell Cycle Proteins/metabolism , Ferroptosis/genetics , Lipid Metabolism/genetics , PPAR alpha/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Cell Cycle Proteins/genetics , Glioblastoma/physiopathology , HCT116 Cells , Humans , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Rats , Tumor Suppressor Protein p53/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
13.
Trends Biochem Sci ; 48(5): 463-476, 2023 05.
Article in English | MEDLINE | ID: mdl-36702698

ABSTRACT

Coenzyme Q (CoQ) is a remarkably hydrophobic, redox-active lipid that empowers diverse cellular processes. Although most known for shuttling electrons between mitochondrial electron transport chain (ETC) complexes, the roles for CoQ are far more wide-reaching and ever-expanding. CoQ serves as a conduit for electrons from myriad pathways to enter the ETC, acts as a cofactor for biosynthetic and catabolic reactions, detoxifies damaging lipid species, and engages in cellular signaling and oxygen sensing. Many open questions remain regarding the biosynthesis, transport, and metabolism of CoQ, which hinders our ability to treat human CoQ deficiency. Here, we recount progress in filling these knowledge gaps, highlight unanswered questions, and underscore the need for novel tools to enable discoveries and improve the treatment of CoQ-related diseases.


Subject(s)
Mitochondrial Diseases , Ubiquinone , Humans , Ubiquinone/metabolism , Mitochondrial Diseases/metabolism , Oxidation-Reduction , Ataxia/metabolism , Lipids
14.
Nature ; 597(7876): 420-425, 2021 09.
Article in English | MEDLINE | ID: mdl-34471290

ABSTRACT

Oxygen is critical for a multitude of metabolic processes that are essential for human life. Biological processes can be identified by treating cells with 18O2 or other isotopically labelled gases and systematically identifying biomolecules incorporating labeled atoms. Here we labelled cell lines of distinct tissue origins with 18O2 to identify the polar oxy-metabolome, defined as polar metabolites labelled with 18O under different physiological O2 tensions. The most highly 18O-labelled feature was 4-hydroxymandelate (4-HMA). We demonstrate that 4-HMA is produced by hydroxyphenylpyruvate dioxygenase-like (HPDL), a protein of previously unknown function in human cells. We identify 4-HMA as an intermediate involved in the biosynthesis of the coenzyme Q10 (CoQ10) headgroup in human cells. The connection of HPDL to CoQ10 biosynthesis provides crucial insights into the mechanisms underlying recently described neurological diseases related to HPDL deficiencies1-4 and cancers with HPDL overexpression5.


Subject(s)
4-Hydroxyphenylpyruvate Dioxygenase/metabolism , Mandelic Acids/metabolism , Metabolome , Ubiquinone/analogs & derivatives , Animals , Cell Line , Female , Humans , Mandelic Acids/analysis , Mice , Mice, Nude , Tyrosine/metabolism , Ubiquinone/biosynthesis
15.
Nature ; 589(7841): 310-314, 2021 01.
Article in English | MEDLINE | ID: mdl-33268896

ABSTRACT

Photosynthetic reaction centres harvest the energy content of sunlight by transporting electrons across an energy-transducing biological membrane. Here we use time-resolved serial femtosecond crystallography1 using an X-ray free-electron laser2 to observe light-induced structural changes in the photosynthetic reaction centre of Blastochloris viridis on a timescale of picoseconds. Structural perturbations first occur at the special pair of chlorophyll molecules of the photosynthetic reaction centre that are photo-oxidized by light. Electron transfer to the menaquinone acceptor on the opposite side of the membrane induces a movement of this cofactor together with lower amplitude protein rearrangements. These observations reveal how proteins use conformational dynamics to stabilize the charge-separation steps of electron-transfer reactions.


Subject(s)
Photosynthetic Reaction Center Complex Proteins/chemistry , Photosynthetic Reaction Center Complex Proteins/metabolism , Bacteriochlorophylls/metabolism , Binding Sites/drug effects , Chlorophyll/metabolism , Chlorophyll/radiation effects , Crystallography , Cytoplasm/metabolism , Electron Transport/drug effects , Electrons , Hyphomicrobiaceae/enzymology , Hyphomicrobiaceae/metabolism , Lasers , Models, Molecular , Oxidation-Reduction/radiation effects , Pheophytins/metabolism , Photosynthetic Reaction Center Complex Proteins/radiation effects , Protons , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism , Vitamin K 2/metabolism
16.
Mol Cell ; 73(4): 763-774.e10, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30661980

ABSTRACT

The biosynthesis of coenzyme Q presents a paradigm for how cells surmount hydrophobic barriers in lipid biology. In eukaryotes, CoQ precursors-among nature's most hydrophobic molecules-must somehow be presented to a series of enzymes peripherally associated with the mitochondrial inner membrane. Here, we reveal that this process relies on custom lipid-binding properties of COQ9. We show that COQ9 repurposes the bacterial TetR fold to bind aromatic isoprenes with high specificity, including CoQ intermediates that likely reside entirely within the bilayer. We reveal a process by which COQ9 associates with cardiolipin-rich membranes and warps the membrane surface to access this cargo. Finally, we identify a molecular interface between COQ9 and the hydroxylase COQ7, motivating a model whereby COQ9 presents intermediates directly to CoQ enzymes. Overall, our results provide a mechanism for how a lipid-binding protein might access, select, and deliver specific cargo from a membrane to promote biosynthesis.


Subject(s)
Membrane Lipids/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Ubiquinone/biosynthesis , Binding Sites , Cardiolipins/metabolism , Crystallography , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship , Tryptophan , Ubiquinone/chemistry , Ubiquinone/genetics
17.
Proc Natl Acad Sci U S A ; 121(13): e2321242121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38507448

ABSTRACT

All biological hydroxylation reactions are thought to derive the oxygen atom from one of three inorganic oxygen donors, O2, H2O2, or H2O. Here, we have identified the organic compound prephenate as the oxygen donor for the three hydroxylation steps of the O2-independent biosynthetic pathway of ubiquinone, a widely distributed lipid coenzyme. Prephenate is an intermediate in the aromatic amino acid pathway and genetic experiments showed that it is essential for ubiquinone biosynthesis in Escherichia coli under anaerobic conditions. Metabolic labeling experiments with 18O-shikimate, a precursor of prephenate, demonstrated the incorporation of 18O atoms into ubiquinone. The role of specific iron-sulfur enzymes belonging to the widespread U32 protein family is discussed. Prephenate-dependent hydroxylation reactions represent a unique biochemical strategy for adaptation to anaerobic environments.


Subject(s)
Cyclohexanecarboxylic Acids , Cyclohexenes , Escherichia coli , Ubiquinone , Hydroxylation , Ubiquinone/metabolism , Escherichia coli/metabolism , Oxygen/metabolism
18.
Proc Natl Acad Sci U S A ; 121(25): e2402384121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38865272

ABSTRACT

Loss of mitochondrial electron transport complex (ETC) function in the retinal pigment epithelium (RPE) in vivo results in RPE dedifferentiation and progressive photoreceptor degeneration, and has been implicated in the pathogenesis of age-related macular degeneration. Xenogenic expression of alternative oxidases in mammalian cells and tissues mitigates phenotypes arising from some mitochondrial electron transport defects, but can exacerbate others. We expressed an alternative oxidase from Ciona intestinalis (AOX) in ETC-deficient murine RPE in vivo to assess the retinal consequences of stimulating coenzyme Q oxidation and respiration without ATP generation. RPE-restricted expression of AOX in this context is surprisingly beneficial. This focused intervention mitigates RPE mTORC1 activation, dedifferentiation, hypertrophy, stress marker expression, pseudohypoxia, and aerobic glycolysis. These RPE cell autonomous changes are accompanied by increased glucose delivery to photoreceptors with attendant improvements in photoreceptor structure and function. RPE-restricted AOX expression normalizes accumulated levels of succinate and 2-hydroxyglutarate in ETC-deficient RPE, and counteracts deficiencies in numerous neural retinal metabolites. These features can be attributed to the activation of mitochondrial inner membrane flavoproteins such as succinate dehydrogenase and proline dehydrogenase, and alleviation of inhibition of 2-oxyglutarate-dependent dioxygenases such as prolyl hydroxylases and epigenetic modifiers. Our work underscores the importance to outer retinal health of coenzyme Q oxidation in the RPE and identifies a metabolic network critical for photoreceptor survival in the context of RPE mitochondrial dysfunction.


Subject(s)
Mitochondria , Oxidoreductases , Plant Proteins , Retinal Pigment Epithelium , Animals , Mitochondria/metabolism , Mice , Oxidoreductases/metabolism , Oxidoreductases/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Plant Proteins/metabolism , Plant Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Ciona intestinalis/metabolism , Ubiquinone/metabolism , Ubiquinone/analogs & derivatives , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/genetics , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology
19.
Nat Chem Biol ; 20(2): 221-233, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37884807

ABSTRACT

Targeting proximity-labeling enzymes to specific cellular locations is a viable strategy for profiling subcellular proteomes. Here, we generated transgenic mice (MAX-Tg) expressing a mitochondrial matrix-targeted ascorbate peroxidase. Comparative analysis of matrix proteomes from the muscle tissues showed differential enrichment of mitochondrial proteins. We found that reticulon 4-interacting protein 1 (RTN4IP1), also known as optic atrophy-10, is enriched in the mitochondrial matrix of muscle tissues and is an NADPH oxidoreductase. Interactome analysis and in vitro enzymatic assays revealed an essential role for RTN4IP1 in coenzyme Q (CoQ) biosynthesis by regulating the O-methylation activity of COQ3. Rtn4ip1-knockout myoblasts had markedly decreased CoQ9 levels and impaired cellular respiration. Furthermore, muscle-specific knockdown of dRtn4ip1 in flies resulted in impaired muscle function, which was reversed by dietary supplementation with soluble CoQ. Collectively, these results demonstrate that RTN4IP1 is a mitochondrial NAD(P)H oxidoreductase essential for supporting mitochondrial respiration activity in the muscle tissue.


Subject(s)
Oxidoreductases , Ubiquinone , Animals , Mice , Drosophila melanogaster , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proteome , Ubiquinone/metabolism , Carrier Proteins
20.
Nature ; 585(7824): 288-292, 2020 09.
Article in English | MEDLINE | ID: mdl-32641834

ABSTRACT

The mitochondrial electron transport chain (ETC) is necessary for tumour growth1-6 and its inhibition has demonstrated anti-tumour efficacy in combination with targeted therapies7-9. Furthermore, human brain and lung tumours display robust glucose oxidation by mitochondria10,11. However, it is unclear why a functional ETC is necessary for tumour growth in vivo. ETC function is coupled to the generation of ATP-that is, oxidative phosphorylation and the production of metabolites by the tricarboxylic acid (TCA) cycle. Mitochondrial complexes I and II donate electrons to ubiquinone, resulting in the generation of ubiquinol and the regeneration of the NAD+ and FAD cofactors, and complex III oxidizes ubiquinol back to ubiquinone, which also serves as an electron acceptor for dihydroorotate dehydrogenase (DHODH)-an enzyme necessary for de novo pyrimidine synthesis. Here we show impaired tumour growth in cancer cells that lack mitochondrial complex III. This phenotype was rescued by ectopic expression of Ciona intestinalis alternative oxidase (AOX)12, which also oxidizes ubiquinol to ubiquinone. Loss of mitochondrial complex I, II or DHODH diminished the tumour growth of AOX-expressing cancer cells deficient in mitochondrial complex III, which highlights the necessity of ubiquinone as an electron acceptor for tumour growth. Cancer cells that lack mitochondrial complex III but can regenerate NAD+ by expression of the NADH oxidase from Lactobacillus brevis (LbNOX)13 targeted to the mitochondria or cytosol were still unable to grow tumours. This suggests that regeneration of NAD+ is not sufficient to drive tumour growth in vivo. Collectively, our findings indicate that tumour growth requires the ETC to oxidize ubiquinol, which is essential to drive the oxidative TCA cycle and DHODH activity.


Subject(s)
Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Ubiquinone/analogs & derivatives , Animals , Cell Line, Tumor , Cell Proliferation , Ciona intestinalis/enzymology , Citric Acid Cycle , Cytosol/metabolism , Dihydroorotate Dehydrogenase , Electron Transport , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Electron Transport Complex III/deficiency , Electron Transport Complex III/metabolism , Humans , Levilactobacillus brevis/enzymology , Male , Mice , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NAD/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Neoplasms/enzymology , Oxidative Phosphorylation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Ubiquinone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL