Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Publication year range
1.
Nat Rev Mol Cell Biol ; 25(1): 65-82, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37773518

ABSTRACT

Mitochondria are multifaceted organelles with key roles in anabolic and catabolic metabolism, bioenergetics, cellular signalling and nutrient sensing, and programmed cell death processes. Their diverse functions are enabled by a sophisticated set of protein components encoded by the nuclear and mitochondrial genomes. The extent and complexity of the mitochondrial proteome remained unclear for decades. This began to change 20 years ago when, driven by the emergence of mass spectrometry-based proteomics, the first draft mitochondrial proteomes were established. In the ensuing decades, further technological and computational advances helped to refine these 'maps', with current estimates of the core mammalian mitochondrial proteome ranging from 1,000 to 1,500 proteins. The creation of these compendia provided a systemic view of an organelle previously studied primarily in a reductionist fashion and has accelerated both basic scientific discovery and the diagnosis and treatment of human disease. Yet numerous challenges remain in understanding mitochondrial biology and translating this knowledge into the medical context. In this Roadmap, we propose a path forward for refining the mitochondrial protein map to enhance its discovery and therapeutic potential. We discuss how emerging technologies can assist the detection of new mitochondrial proteins, reveal their patterns of expression across diverse tissues and cell types, and provide key information on proteoforms. We highlight the power of an enhanced map for systematically defining the functions of its members. Finally, we examine the utility of an expanded, functionally annotated mitochondrial proteome in a translational setting for aiding both diagnosis of mitochondrial disease and targeting of mitochondria for treatment.


Subject(s)
Mitochondrial Diseases , Proteome , Animals , Humans , Proteome/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Organelles/metabolism , Mitochondrial Diseases/metabolism , Mass Spectrometry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mammals/metabolism
2.
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
3.
Cell ; 161(7): 1619-32, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091039

ABSTRACT

The existence of extracellular phosphoproteins has been acknowledged for over a century. However, research in this area has been undeveloped largely because the kinases that phosphorylate secreted proteins have escaped identification. Fam20C is a kinase that phosphorylates S-x-E/pS motifs on proteins in milk and in the extracellular matrix of bones and teeth. Here, we show that Fam20C generates the majority of the extracellular phosphoproteome. Using CRISPR/Cas9 genome editing, mass spectrometry, and biochemistry, we identify more than 100 secreted phosphoproteins as genuine Fam20C substrates. Further, we show that Fam20C exhibits broader substrate specificity than previously appreciated. Functional annotations of Fam20C substrates suggest roles for the kinase beyond biomineralization, including lipid homeostasis, wound healing, and cell migration and adhesion. Our results establish Fam20C as the major secretory pathway protein kinase and serve as a foundation for new areas of investigation into the role of secreted protein phosphorylation in human biology and disease.


Subject(s)
Casein Kinase I/chemistry , Casein Kinase I/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Amino Acid Sequence , Blood Proteins/metabolism , Casein Kinase I/genetics , Cell Adhesion , Cell Movement , Cerebrospinal Fluid Proteins/metabolism , Extracellular Matrix Proteins/genetics , Gene Knockout Techniques , Gene Ontology , Humans , Molecular Sequence Data , Phosphoproteins/analysis , Secretory Pathway , Substrate Specificity
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 ; 606(7913): 382-388, 2022 06.
Article in English | MEDLINE | ID: mdl-35614220

ABSTRACT

Mitochondria are epicentres of eukaryotic metabolism and bioenergetics. Pioneering efforts in recent decades have established the core protein componentry of these organelles1 and have linked their dysfunction to more than 150 distinct disorders2,3. Still, hundreds of mitochondrial proteins lack clear functions4, and the underlying genetic basis for approximately 40% of mitochondrial disorders remains unresolved5. Here, to establish a more complete functional compendium of human mitochondrial proteins, we profiled more than 200 CRISPR-mediated HAP1 cell knockout lines using mass spectrometry-based multiomics analyses. This effort generated approximately 8.3 million distinct biomolecule measurements, providing a deep survey of the cellular responses to mitochondrial perturbations and laying a foundation for mechanistic investigations into protein function. Guided by these data, we discovered that PIGY upstream open reading frame (PYURF) is an S-adenosylmethionine-dependent methyltransferase chaperone that supports both complex I assembly and coenzyme Q biosynthesis and is disrupted in a previously unresolved multisystemic mitochondrial disorder. We further linked the putative zinc transporter SLC30A9 to mitochondrial ribosomes and OxPhos integrity and established RAB5IF as the second gene harbouring pathogenic variants that cause cerebrofaciothoracic dysplasia. Our data, which can be explored through the interactive online MITOMICS.app resource, suggest biological roles for many other orphan mitochondrial proteins that still lack robust functional characterization and define a rich cell signature of mitochondrial dysfunction that can support the genetic diagnosis of mitochondrial diseases.


Subject(s)
Mitochondria , Mitochondrial Proteins , Cation Transport Proteins , Cell Cycle Proteins , Energy Metabolism , Humans , Mass Spectrometry , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Transcription Factors , rab5 GTP-Binding Proteins
6.
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
7.
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
8.
J Biol Chem ; 300(5): 107269, 2024 May.
Article in English | MEDLINE | ID: mdl-38588811

ABSTRACT

Coenzyme Q10 (CoQ10) is an important cofactor and antioxidant for numerous cellular processes, and its deficiency has been linked to human disorders including mitochondrial disease, heart failure, Parkinson's disease, and hypertension. Unfortunately, treatment with exogenous CoQ10 is often ineffective, likely due to its extreme hydrophobicity and high molecular weight. Here, we show that less hydrophobic CoQ species with shorter isoprenoid tails can serve as viable substitutes for CoQ10 in human cells. We demonstrate that CoQ4 can perform multiple functions of CoQ10 in CoQ-deficient cells at markedly lower treatment concentrations, motivating further investigation of CoQ4 as a supplement for CoQ10 deficiencies. In addition, we describe the synthesis and evaluation of an initial set of compounds designed to target CoQ4 selectively to mitochondria using triphenylphosphonium. Our results indicate that select versions of these compounds can successfully be delivered to mitochondria in a cell model and be cleaved to produce CoQ4, laying the groundwork for further development.


Subject(s)
Ataxia , Mitochondria , Mitochondrial Diseases , Muscle Weakness , Ubiquinone , Humans , Mitochondria/enzymology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Muscle Weakness/enzymology , Muscle Weakness/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Hep G2 Cells
9.
Nat Chem Biol ; 19(3): 265-274, 2023 03.
Article in English | MEDLINE | ID: mdl-36266351

ABSTRACT

Pyruvate dehydrogenase complex (PDHC) and oxoglutarate dehydrogenase complex (OGDC), which belong to the mitochondrial α-ketoacid dehydrogenase family, play crucial roles in cellular metabolism. These multi-subunit enzyme complexes use lipoic arms covalently attached to their E2 subunits to transfer an acyl group to coenzyme A (CoA). Here, we report a novel mechanism capable of substantially inhibiting PDHC and OGDC: reactive nitrogen species (RNS) can covalently modify the thiols on their lipoic arms, generating a series of adducts that block catalytic activity. S-Nitroso-CoA, a product between RNS and the E2 subunit's natural substrate, CoA, can efficiently deliver these modifications onto the lipoic arm. We found RNS-mediated inhibition of PDHC and OGDC occurs during classical macrophage activation, driving significant rewiring of cellular metabolism over time. This work provides a new mechanistic link between RNS and mitochondrial metabolism with potential relevance for numerous physiological and pathological conditions in which RNS accumulate.


Subject(s)
Arm , Nitric Oxide , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) , Pyruvate Dehydrogenase Complex/metabolism , Multienzyme Complexes
10.
Nat Chem Biol ; 19(2): 230-238, 2023 02.
Article in English | MEDLINE | ID: mdl-36302899

ABSTRACT

Small-molecule tools have enabled mechanistic investigations and therapeutic targeting of the protein kinase-like (PKL) superfamily. However, such tools are still lacking for many PKL members, including the highly conserved and disease-related UbiB family. Here, we sought to develop and characterize an inhibitor for the archetypal UbiB member COQ8, whose function is essential for coenzyme Q (CoQ) biosynthesis. Guided by crystallography, activity assays and cellular CoQ measurements, we repurposed the 4-anilinoquinoline scaffold to selectively inhibit human COQ8A in cells. Our chemical tool promises to lend mechanistic insights into the activities of these widespread and understudied proteins and to offer potential therapeutic strategies for human diseases connected to their dysfunction.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Saccharomyces cerevisiae/metabolism , Ubiquinone/pharmacology , Ubiquinone/chemistry , Saccharomyces cerevisiae Proteins/metabolism
11.
Mol Cell ; 68(5): 970-977.e11, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29220658

ABSTRACT

Mitoproteases are becoming recognized as key regulators of diverse mitochondrial functions, although their direct substrates are often difficult to discern. Through multi-omic profiling of diverse Saccharomyces cerevisiae mitoprotease deletion strains, we predicted numerous associations between mitoproteases and distinct mitochondrial processes. These include a strong association between the mitochondrial matrix octapeptidase Oct1p and coenzyme Q (CoQ) biosynthesis-a pathway essential for mitochondrial respiration. Through Edman sequencing and in vitro and in vivo biochemistry, we demonstrated that Oct1p directly processes the N terminus of the CoQ-related methyltransferase, Coq5p, which markedly improves its stability. A single mutation to the Oct1p recognition motif in Coq5p disrupted its processing in vivo, leading to CoQ deficiency and respiratory incompetence. This work defines the Oct1p processing of Coq5p as an essential post-translational event for proper CoQ production. Additionally, our data visualization tool enables efficient exploration of mitoprotease profiles that can serve as the basis for future mechanistic investigations.


Subject(s)
Aminopeptidases/metabolism , Energy Metabolism , Metabolomics/methods , Methyltransferases/metabolism , Mitochondria/enzymology , Proteomics/methods , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Ubiquinone/biosynthesis , Aminopeptidases/genetics , Enzyme Stability , Genotype , Methyltransferases/genetics , Mutation , Phenotype , Protein Domains , Protein Processing, Post-Translational , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Time Factors , Ubiquinone/genetics
12.
J Biol Chem ; 299(3): 102981, 2023 03.
Article in English | MEDLINE | ID: mdl-36739946

ABSTRACT

Chalcone isomerases (CHIs) have well-established roles in the biosynthesis of plant flavonoid metabolites. Saccharomyces cerevisiae possesses two predicted CHI-like proteins, Aim18p (encoded by YHR198C) and Aim46p (YHR199C), but it lacks other enzymes of the flavonoid pathway, suggesting that Aim18p and Aim46p employ the CHI fold for distinct purposes. Here, we demonstrate using proteinase K protection assays, sodium carbonate extractions, and crystallography that Aim18p and Aim46p reside on the mitochondrial inner membrane and adopt CHI folds, but they lack select active site residues and possess an extra fungal-specific loop. Consistent with these differences, Aim18p and Aim46p lack CHI activity and also the fatty acid-binding capabilities of other CHI-like proteins, but instead bind heme. We further show that diverse fungal homologs also bind heme and that Aim18p and Aim46p possess structural homology to a bacterial hemoprotein. Collectively, our work reveals a distinct function and cellular localization for two CHI-like proteins, introduces a new variation of a hemoprotein fold, and suggests that ancestral CHI-like proteins were hemoproteins.


Subject(s)
Intramolecular Lyases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Flavonoids/metabolism , Intramolecular Lyases/chemistry , Intramolecular Lyases/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
13.
Mol Cell ; 63(4): 621-632, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499296

ABSTRACT

Mitochondria are essential for numerous cellular processes, yet hundreds of their proteins lack robust functional annotation. To reveal functions for these proteins (termed MXPs), we assessed condition-specific protein-protein interactions for 50 select MXPs using affinity enrichment mass spectrometry. Our data connect MXPs to diverse mitochondrial processes, including multiple aspects of respiratory chain function. Building upon these observations, we validated C17orf89 as a complex I (CI) assembly factor. Disruption of C17orf89 markedly reduced CI activity, and its depletion is found in an unresolved case of CI deficiency. We likewise discovered that LYRM5 interacts with and deflavinates the electron-transferring flavoprotein that shuttles electrons to coenzyme Q (CoQ). Finally, we identified a dynamic human CoQ biosynthetic complex involving multiple MXPs whose topology we map using purified components. Collectively, our data lend mechanistic insight into respiratory chain-related activities and prioritize hundreds of additional interactions for further exploration of mitochondrial protein function.


Subject(s)
Electron Transport Chain Complex Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Proteomics/methods , Databases, Protein , Electron Transport Chain Complex Proteins/genetics , Electron Transport Complex I/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Mitochondrial Proteins/genetics , RNA Interference , Signal Transduction , Transfection , Ubiquinone/metabolism
14.
Mol Cell ; 63(4): 608-620, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499294

ABSTRACT

The UbiB protein kinase-like (PKL) family is widespread, comprising one-quarter of microbial PKLs and five human homologs, yet its biochemical activities remain obscure. COQ8A (ADCK3) is a mammalian UbiB protein associated with ubiquinone (CoQ) biosynthesis and an ataxia (ARCA2) through unclear means. We show that mice lacking COQ8A develop a slowly progressive cerebellar ataxia linked to Purkinje cell dysfunction and mild exercise intolerance, recapitulating ARCA2. Interspecies biochemical analyses show that COQ8A and yeast Coq8p specifically stabilize a CoQ biosynthesis complex through unorthodox PKL functions. Although COQ8 was predicted to be a protein kinase, we demonstrate that it lacks canonical protein kinase activity in trans. Instead, COQ8 has ATPase activity and interacts with lipid CoQ intermediates, functions that are likely conserved across all domains of life. Collectively, our results lend insight into the molecular activities of the ancient UbiB family and elucidate the biochemical underpinnings of a human disease.


Subject(s)
Behavior, Animal , Cerebellar Ataxia/enzymology , Cerebellum/enzymology , Mitochondrial Proteins/deficiency , Muscle, Skeletal/enzymology , Ubiquinone/deficiency , Animals , COS Cells , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Cerebellar Ataxia/psychology , Cerebellum/physiopathology , Cerebellum/ultrastructure , Chlorocebus aethiops , Disease Models, Animal , Exercise Tolerance , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Lipid Metabolism , Male , Maze Learning , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Models, Molecular , Motor Activity , Muscle Strength , Muscle, Skeletal/physiopathology , Phenotype , Protein Binding , Protein Conformation , Proteomics/methods , Recognition, Psychology , Rotarod Performance Test , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Seizures/enzymology , Seizures/genetics , Seizures/physiopathology , Structure-Activity Relationship , Time Factors , Transfection , Ubiquinone/chemistry , Ubiquinone/genetics
15.
Am J Hum Genet ; 106(1): 92-101, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31866046

ABSTRACT

Leigh syndrome is one of the most common neurological phenotypes observed in pediatric mitochondrial disease presentations. It is characterized by symmetrical lesions found on neuroimaging in the basal ganglia, thalamus, and brainstem and by a loss of motor skills and delayed developmental milestones. Genetic diagnosis of Leigh syndrome is complicated on account of the vast genetic heterogeneity with >75 candidate disease-associated genes having been reported to date. Candidate genes are still emerging, being identified when "omics" tools (genomics, proteomics, and transcriptomics) are applied to manipulated cell lines and cohorts of clinically characterized individuals who lack a genetic diagnosis. NDUFAF8 is one such protein; it has been found to interact with the well-characterized complex I (CI) assembly factor NDUFAF5 in a large-scale protein-protein interaction screen. Diagnostic next-generation sequencing has identified three unrelated pediatric subjects, each with a clinical diagnosis of Leigh syndrome, who harbor bi-allelic pathogenic variants in NDUFAF8. These variants include a recurrent splicing variant that was initially overlooked due to its deep-intronic location. Subject fibroblasts were found to express a complex I deficiency, and lentiviral transduction with wild-type NDUFAF8-cDNA ameliorated both the assembly defect and the biochemical deficiency. Complexome profiling of subject fibroblasts demonstrated a complex I assembly defect, and the stalled assembly intermediates corroborate the role of NDUFAF8 in early complex I assembly. This report serves to expand the genetic heterogeneity associated with Leigh syndrome and to validate the clinical utility of orphan protein characterization. We also highlight the importance of evaluating intronic sequence when a single, definitively pathogenic variant is identified during diagnostic testing.


Subject(s)
Electron Transport Complex I/deficiency , Fibroblasts/pathology , Leigh Disease/etiology , Mitochondrial Diseases/etiology , Mitochondrial Proteins/genetics , Mutation , NADH Dehydrogenase/genetics , Alleles , Female , Fibroblasts/metabolism , Humans , Infant , Leigh Disease/pathology , Male , Mitochondrial Diseases/pathology , Pedigree , Phenotype
16.
Nat Methods ; 17(12): 1222-1228, 2020 12.
Article in English | MEDLINE | ID: mdl-33230323

ABSTRACT

Liquid chromatography-mass spectrometry (LC-MS) delivers sensitive peptide analysis for proteomics but requires extensive analysis time, reducing throughput. Here, we demonstrate that gas-phase peptide separation instead of LC enables fast proteome analysis. Using direct infusion-shotgun proteome analysis (DI-SPA) by data-independent acquisition mass spectrometry (DIA-MS), we demonstrate the targeted quantification of over 500 proteins within minutes of MS data collection (~3.5 proteins per second). We show the utility of this technology in performing a complex multifactorial proteomic study of interactions between nutrients, genotype and mitochondrial toxins in a collection of cultured human cells. More than 45,000 quantitative protein measurements from 132 samples were achieved in only ~4.4 h of MS data collection. Enabling fast, unbiased proteome quantification without LC, DI-SPA offers an approach to boost throughput, critical to drug and biomarker discovery studies that require analysis of thousands of proteomes.


Subject(s)
Gas Chromatography-Mass Spectrometry/methods , Proteome/analysis , Proteomics/methods , A549 Cells , Cell Line, Tumor , Gene Expression Profiling/methods , HEK293 Cells , Humans , MCF-7 Cells
17.
Cell ; 134(1): 112-23, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18614015

ABSTRACT

Mitochondria are complex organelles whose dysfunction underlies a broad spectrum of human diseases. Identifying all of the proteins resident in this organelle and understanding how they integrate into pathways represent major challenges in cell biology. Toward this goal, we performed mass spectrometry, GFP tagging, and machine learning to create a mitochondrial compendium of 1098 genes and their protein expression across 14 mouse tissues. We link poorly characterized proteins in this inventory to known mitochondrial pathways by virtue of shared evolutionary history. Using this approach, we predict 19 proteins to be important for the function of complex I (CI) of the electron transport chain. We validate a subset of these predictions using RNAi, including C8orf38, which we further show harbors an inherited mutation in a lethal, infantile CI deficiency. Our results have important implications for understanding CI function and pathogenesis and, more generally, illustrate how our compendium can serve as a foundation for systematic investigations of mitochondria.


Subject(s)
Leigh Disease/genetics , Mitochondria/chemistry , Mitochondrial Proteins/analysis , Proteome , Animals , Databases, Protein , Electron Transport Complex I/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Organ Specificity
18.
Mol Cell ; 57(1): 83-94, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25498144

ABSTRACT

The ancient UbiB protein kinase-like family is involved in isoprenoid lipid biosynthesis and is implicated in human diseases, but demonstration of UbiB kinase activity has remained elusive for unknown reasons. Here, we quantitatively define UbiB-specific sequence motifs and reveal their positions within the crystal structure of a UbiB protein, ADCK3. We find that multiple UbiB-specific features are poised to inhibit protein kinase activity, including an N-terminal domain that occupies the typical substrate binding pocket and a unique A-rich loop that limits ATP binding by establishing an unusual selectivity for ADP. A single alanine-to-glycine mutation of this loop flips this coenzyme selectivity and enables autophosphorylation but inhibits coenzyme Q biosynthesis in vivo, demonstrating functional relevance for this unique feature. Our work provides mechanistic insight into UbiB enzyme activity and establishes a molecular foundation for further investigation of how UbiB family proteins affect diseases and diverse biological pathways.


Subject(s)
Mitochondria/chemistry , Mitochondrial Proteins/chemistry , Ubiquinone/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Phosphorylation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Ubiquinone/biosynthesis
19.
J Biol Chem ; 297(1): 100880, 2021 07.
Article in English | MEDLINE | ID: mdl-34144036

ABSTRACT

More than half a century ago, reversible protein phosphorylation was linked to mitochondrial metabolism through the regulation of pyruvate dehydrogenase. Since this discovery, the number of identified mitochondrial protein phosphorylation sites has increased by orders of magnitude, driven largely by technological advances in mass spectrometry-based phosphoproteomics. However, the majority of these modifications remain uncharacterized, rendering their function and relevance unclear. Nonetheless, recent studies have shown that disruption of resident mitochondrial protein phosphatases causes substantial metabolic dysfunction across organisms, suggesting that proper management of mitochondrial phosphorylation is vital for organellar and organismal homeostasis. While these data suggest that phosphorylation within mitochondria is of critical importance, significant gaps remain in our knowledge of how these modifications influence organellar function. Here, we curate publicly available datasets to map the extent of protein phosphorylation within mammalian mitochondria and to highlight the known functions of mitochondrial-resident phosphatases. We further propose models by which phosphorylation may affect mitochondrial enzyme activities, protein import and processing, and overall organellar homeostasis.


Subject(s)
Mitochondrial Proteins/metabolism , Phosphoproteins/metabolism , Proteome/metabolism , Animals , Humans , Mitochondrial Proteins/genetics , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Proteome/genetics
20.
J Biol Chem ; 296: 100643, 2021.
Article in English | MEDLINE | ID: mdl-33862086

ABSTRACT

Coenzyme Q (CoQ), a redox-active lipid essential for oxidative phosphorylation, is synthesized by virtually all cells, but how eukaryotes make the universal CoQ head group precursor 4-hydroxybenzoate (4-HB) from tyrosine is unknown. The first and last steps of this pathway have been defined in Saccharomyces cerevisiae, but the intermediates and enzymes involved in converting 4-hydroxyphenylpyruvate (4-HPP) to 4-hydroxybenzaldehyde (4-HBz) have not been described. Here, we interrogate this pathway with genetic screens, targeted LC-MS, and chemical genetics. We identify three redundant aminotransferases (Bna3, Bat2, and Aat2) that support CoQ biosynthesis in the absence of the established pathway tyrosine aminotransferases, Aro8 and Aro9. We use isotope labeling to identify bona fide tyrosine catabolites, including 4-hydroxyphenylacetate (4-HPA) and 4-hydroxyphenyllactate (4-HPL). Additionally, we find multiple compounds that rescue this pathway when exogenously supplemented, most notably 4-hydroxyphenylacetaldehyde (4-HPAA) and 4-hydroxymandelate (4-HMA). Finally, we show that the Ehrlich pathway decarboxylase Aro10 is dispensable for 4-HB production. These results define new features of 4-HB synthesis in yeast, demonstrate the redundant nature of this pathway, and provide a foundation for further study.


Subject(s)
Parabens/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transaminases/metabolism , Tyrosine/metabolism , Ubiquinone/analogs & derivatives , Oxidation-Reduction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Transaminases/genetics , Ubiquinone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL