Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.298
Filter
1.
Cell ; 187(10): 2359-2374.e18, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38653240

ABSTRACT

Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.


Subject(s)
Adipose Tissue, Brown , Amino Acids, Branched-Chain , Insulin Resistance , Mitochondria , Nitrogen , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Amino Acids, Branched-Chain/metabolism , Mice , Nitrogen/metabolism , Mitochondria/metabolism , Male , Humans , Energy Metabolism , Mice, Inbred C57BL , Oxidative Stress , Insulin/metabolism , Diet, High-Fat , Adipocytes, Brown/metabolism , Signal Transduction
2.
Annu Rev Biochem ; 90: 535-558, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33556281

ABSTRACT

Members of the mitochondrial carrier family [solute carrier family 25 (SLC25)] transport nucleotides, amino acids, carboxylic acids, fatty acids, inorganic ions, and vitamins across the mitochondrial inner membrane. They are important for many cellular processes, such as oxidative phosphorylation of lipids and sugars, amino acid metabolism, macromolecular synthesis, ion homeostasis, cellular regulation, and differentiation. Here, we describe the functional elements of the transport mechanism of mitochondrial carriers, consisting of one central substrate-binding site and two gates with salt-bridge networks on either side of the carrier. Binding of the substrate during import causes three gate elements to rotate inward, forming the cytoplasmic network and closing access to the substrate-binding site from the intermembrane space. Simultaneously, three core elements rock outward, disrupting the matrix network and opening the substrate-binding site to the matrix side of the membrane. During export, substrate binding triggers conformational changes involving the same elements but operating in reverse.


Subject(s)
Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Aggrecans/chemistry , Aggrecans/genetics , Aggrecans/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Binding Sites , Biological Transport , Calcium/metabolism , Cardiolipins/metabolism , Conserved Sequence , Cytoplasm/metabolism , Humans , Mitochondrial ADP, ATP Translocases/chemistry , Mitochondrial ADP, ATP Translocases/metabolism , Mutation , Protein Conformation , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
3.
Cell ; 182(6): 1490-1507.e19, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32916131

ABSTRACT

Metabolic reprogramming is a key feature of many cancers, but how and when it contributes to tumorigenesis remains unclear. Here we demonstrate that metabolic reprogramming induced by mitochondrial fusion can be rate-limiting for immortalization of tumor-initiating cells (TICs) and trigger their irreversible dedication to tumorigenesis. Using single-cell transcriptomics, we find that Drosophila brain tumors contain a rapidly dividing stem cell population defined by upregulation of oxidative phosphorylation (OxPhos). We combine targeted metabolomics and in vivo genetic screening to demonstrate that OxPhos is required for tumor cell immortalization but dispensable in neural stem cells (NSCs) giving rise to tumors. Employing an in vivo NADH/NAD+ sensor, we show that NSCs precisely increase OxPhos during immortalization. Blocking OxPhos or mitochondrial fusion stalls TICs in quiescence and prevents tumorigenesis through impaired NAD+ regeneration. Our work establishes a unique connection between cellular metabolism and immortalization of tumor-initiating cells.


Subject(s)
Brain Neoplasms/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Mitochondrial Dynamics , NAD/metabolism , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Oxidative Phosphorylation , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Citric Acid Cycle/genetics , Computational Biology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Glycolysis/genetics , Mass Spectrometry , Metabolomics , Microscopy, Electron, Transmission , Multigene Family , Neural Stem Cells/pathology , Oxygen Consumption/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Single-Cell Analysis , Transcriptome/genetics
4.
Annu Rev Biochem ; 88: 59-83, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30830799

ABSTRACT

Directional transport of protons across an energy transducing membrane-proton pumping-is ubiquitous in biology. Bacteriorhodopsin (bR) is a light-driven proton pump that is activated by a buried all-trans retinal chromophore being photoisomerized to a 13-cis conformation. The mechanism by which photoisomerization initiates directional proton transport against a proton concentration gradient has been studied by a myriad of biochemical, biophysical, and structural techniques. X-ray free electron lasers (XFELs) have created new opportunities to probe the structural dynamics of bR at room temperature on timescales from femtoseconds to milliseconds using time-resolved serial femtosecond crystallography (TR-SFX). Wereview these recent developments and highlight where XFEL studies reveal new details concerning the structural mechanism of retinal photoisomerization and proton pumping. We also discuss the extent to which these insights were anticipated by earlier intermediate trapping studies using synchrotron radiation. TR-SFX will open up the field for dynamical studies of other proteins that are not naturally light-sensitive.


Subject(s)
Bacteriorhodopsins/ultrastructure , Lasers , Protons , Retinaldehyde/chemistry , X-Ray Diffraction/methods , Bacteriorhodopsins/chemistry , Bacteriorhodopsins/metabolism , Crystallography/instrumentation , Crystallography/methods , Halobacterium salinarum/chemistry , Halobacterium salinarum/metabolism , Ion Transport , Models, Molecular , Protein Conformation , Retinaldehyde/metabolism , Synchrotrons/instrumentation , X-Rays
5.
Cell ; 179(5): 1098-1111.e23, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730852

ABSTRACT

We report a 100-million atom-scale model of an entire cell organelle, a photosynthetic chromatophore vesicle from a purple bacterium, that reveals the cascade of energy conversion steps culminating in the generation of ATP from sunlight. Molecular dynamics simulations of this vesicle elucidate how the integral membrane complexes influence local curvature to tune photoexcitation of pigments. Brownian dynamics of small molecules within the chromatophore probe the mechanisms of directional charge transport under various pH and salinity conditions. Reproducing phenotypic properties from atomistic details, a kinetic model evinces that low-light adaptations of the bacterium emerge as a spontaneous outcome of optimizing the balance between the chromatophore's structural integrity and robust energy conversion. Parallels are drawn with the more universal mitochondrial bioenergetic machinery, from whence molecular-scale insights into the mechanism of cellular aging are inferred. Together, our integrative method and spectroscopic experiments pave the way to first-principles modeling of whole living cells.


Subject(s)
Cells/metabolism , Energy Metabolism , Adaptation, Physiological/radiation effects , Adenosine Triphosphate/metabolism , Benzoquinones/metabolism , Cell Membrane/metabolism , Cell Membrane/radiation effects , Cells/radiation effects , Chromatophores/metabolism , Cytochromes c2/metabolism , Diffusion , Electron Transport/radiation effects , Energy Metabolism/radiation effects , Environment , Hydrogen Bonding , Kinetics , Light , Molecular Dynamics Simulation , Phenotype , Proteins/metabolism , Rhodobacter sphaeroides/physiology , Rhodobacter sphaeroides/radiation effects , Static Electricity , Stress, Physiological/radiation effects , Temperature
6.
Cell ; 176(3): 435-447.e15, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30611538

ABSTRACT

Mitochondrial ADP/ATP carriers transport ADP into the mitochondrial matrix for ATP synthesis, and ATP out to fuel the cell, by cycling between cytoplasmic-open and matrix-open states. The structure of the cytoplasmic-open state is known, but it has proved difficult to understand the transport mechanism in the absence of a structure in the matrix-open state. Here, we describe the structure of the matrix-open state locked by bongkrekic acid bound in the ADP/ATP-binding site at the bottom of the central cavity. The cytoplasmic side of the carrier is closed by conserved hydrophobic residues, and a salt bridge network, braced by tyrosines. Glycine and small amino acid residues allow close-packing of helices on the matrix side. Uniquely, the carrier switches between states by rotation of its three domains about a fulcrum provided by the substrate-binding site. Because these features are highly conserved, this mechanism is likely to apply to the whole mitochondrial carrier family. VIDEO ABSTRACT.


Subject(s)
Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial ADP, ATP Translocases/ultrastructure , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , Biological Transport , Bongkrekic Acid/metabolism , Cytoplasm/metabolism , Mitochondria/physiology , Mitochondrial ADP, ATP Translocases/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Membrane Transport Proteins/ultrastructure , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
7.
Physiol Rev ; 103(4): 2451-2506, 2023 10 01.
Article in English | MEDLINE | ID: mdl-36996412

ABSTRACT

Chronic kidney disease (CKD) affects >10% of the world population, with increasing prevalence in middle age. The risk for CKD is dependent on the number of functioning nephrons through the life cycle, and 50% of nephrons are lost through normal aging, revealing their vulnerability to internal and external stressors. Factors responsible for CKD remain poorly understood, with limited availability of biomarkers or effective therapy to slow progression. This review draws on the disciplines of evolutionary medicine and bioenergetics to account for the heterogeneous nephron injury that characterizes progressive CKD following episodes of acute kidney injury with incomplete recovery. The evolution of symbiosis in eukaryotes led to the efficiencies of oxidative phosphorylation and the rise of metazoa. Adaptations to ancestral environments are the products of natural selection that have shaped the mammalian nephron with its vulnerabilities to ischemic, hypoxic, and toxic injury. Reproductive fitness rather than longevity has served as the driver of evolution, constrained by available energy and its allocation to homeostatic responses through the life cycle. Metabolic plasticity has evolved in parallel with robustness necessary to preserve complex developmental programs, and adaptations that optimize survival through reproductive years can become maladaptive with aging, reflecting antagonistic pleiotropy. Consequently, environmental stresses promote trade-offs and mismatches that result in cell fate decisions that ultimately lead to nephron loss. Elucidation of the bioenergetic adaptations by the nephron to ancestral and contemporary environments may lead to the development of new biomarkers of kidney disease and new therapies to reduce the global burden of progressive CKD.


Subject(s)
Kidney , Renal Insufficiency, Chronic , Middle Aged , Animals , Humans , Kidney/metabolism , Nephrons/metabolism , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/metabolism , Aging , Energy Metabolism , Mammals
8.
Trends Biochem Sci ; 49(6): 506-519, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565497

ABSTRACT

In mitochondria, the oxidation of nutrients is coupled to ATP synthesis by the generation of a protonmotive force across the mitochondrial inner membrane. In mammalian brown adipose tissue (BAT), uncoupling protein 1 (UCP1, SLC25A7), a member of the SLC25 mitochondrial carrier family, dissipates the protonmotive force by facilitating the return of protons to the mitochondrial matrix. This process short-circuits the mitochondrion, generating heat for non-shivering thermogenesis. Recent cryo-electron microscopy (cryo-EM) structures of human UCP1 have provided new molecular insights into the inhibition and activation of thermogenesis. Here, we discuss these structures, describing how purine nucleotides lock UCP1 in a proton-impermeable conformation and rationalizing potential conformational changes of this carrier in response to fatty acid activators that enable proton leak for thermogenesis.


Subject(s)
Thermogenesis , Uncoupling Protein 1 , Humans , Uncoupling Protein 1/metabolism , Animals , Mitochondria/metabolism , Adipose Tissue, Brown/metabolism
9.
EMBO J ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937634

ABSTRACT

Members of the SLC25 mitochondrial carrier family link cytosolic and mitochondrial metabolism and support cellular maintenance and growth by transporting compounds across the mitochondrial inner membrane. Their monomeric or dimeric state and kinetic mechanism have been a matter of long-standing debate. It is believed by some that they exist as homodimers and transport substrates with a sequential kinetic mechanism, forming a ternary complex where both exchanged substrates are bound simultaneously. Some studies, in contrast, have provided evidence indicating that the mitochondrial ADP/ATP carrier (SLC25A4) functions as a monomer, has a single substrate binding site, and operates with a ping-pong kinetic mechanism, whereby ADP is imported before ATP is exported. Here we reanalyze the oligomeric state and kinetic properties of the human mitochondrial citrate carrier (SLC25A1), dicarboxylate carrier (SLC25A10), oxoglutarate carrier (SLC25A11), and aspartate/glutamate carrier (SLC25A13), all previously reported to be dimers with a sequential kinetic mechanism. We demonstrate that they are monomers, except for dimeric SLC25A13, and operate with a ping-pong kinetic mechanism in which the substrate import and export steps occur consecutively. These observations are consistent with a common transport mechanism, based on a functional monomer, in which a single central substrate-binding site is alternately accessible.

10.
Trends Biochem Sci ; 48(11): 920-922, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37704489

ABSTRACT

Mitochondrial dysfunction is a central hallmark of aging and energy transduction is a promising target for longevity interventions. New research suggests that interventions in how energy is transduced could benefit healthy longevity. Here, we propose using light as an alternative energy source to fuel mitochondria and increase metazoan lifespan.

11.
Mol Cell ; 75(6): 1131-1146.e6, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31492636

ABSTRACT

The mitochondrial electron transport chain complexes are organized into supercomplexes (SCs) of defined stoichiometry, which have been proposed to regulate electron flux via substrate channeling. We demonstrate that CoQ trapping in the isolated SC I+III2 limits complex (C)I turnover, arguing against channeling. The SC structure, resolved at up to 3.8 Å in four distinct states, suggests that CoQ oxidation may be rate limiting because of unequal access of CoQ to the active sites of CIII2. CI shows a transition between "closed" and "open" conformations, accompanied by the striking rotation of a key transmembrane helix. Furthermore, the state of CI affects the conformational flexibility within CIII2, demonstrating crosstalk between the enzymes. CoQ was identified at only three of the four binding sites in CIII2, suggesting that interaction with CI disrupts CIII2 symmetry in a functionally relevant manner. Together, these observations indicate a more nuanced functional role for the SCs.


Subject(s)
Electron Transport Complex III/chemistry , Electron Transport Complex I/chemistry , Mitochondria, Heart/enzymology , Animals , Crystallography, X-Ray , Protein Structure, Quaternary , Sheep
12.
Proc Natl Acad Sci U S A ; 121(9): e2308796121, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38386708

ABSTRACT

Noise control, together with other regulatory functions facilitated by microRNAs (miRNAs), is believed to have played important roles in the evolution of multicellular eukaryotic organisms. miRNAs can dampen protein fluctuations via enhanced degradation of messenger RNA (mRNA), but this requires compensation by increased mRNA transcription to maintain the same expression levels. The overall mechanism is metabolically expensive, leading to questions about how it might have evolved in the first place. We develop a stochastic model of miRNA noise regulation, coupled with a detailed analysis of the associated metabolic costs. Additionally, we calculate binding free energies for a range of miRNA seeds, the short sequences which govern target recognition. We argue that natural selection may have fine-tuned the Michaelis-Menten constant [Formula: see text] describing miRNA-mRNA affinity and show supporting evidence from analysis of experimental data. [Formula: see text] is constrained by seed length, and optimal noise control (minimum protein variance at a given energy cost) is achievable for seeds of 6 to 7 nucleotides in length, the most commonly observed types. Moreover, at optimality, the degree of noise reduction approaches the theoretical bound set by the Wiener-Kolmogorov linear filter. The results illustrate how selective pressure toward energy efficiency has potentially shaped a crucial regulatory pathway in eukaryotes.


Subject(s)
Eukaryota , MicroRNAs , MicroRNAs/genetics , Mutant Proteins , RNA, Messenger , Energy Metabolism/genetics
13.
Proc Natl Acad Sci U S A ; 121(12): e2319473121, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38478695

ABSTRACT

Hydrogen sulfide exposure in moderate doses can induce profound but reversible hypometabolism in mammals. At a cellular level, H2S inhibits the electron transport chain (ETC), augments aerobic glycolysis, and glutamine-dependent carbon utilization via reductive carboxylation; however, the durability of these changes is unknown. We report that despite its volatility, H2S preconditioning increases P50(O2), the O2 pressure for half-maximal cellular respiration, and has pleiotropic effects on oxidative metabolism that persist up to 24 to 48 h later. Notably, cyanide, another complex IV inhibitor, does not induce this type of metabolic memory. Sulfide-mediated prolonged fractional inhibition of complex IV by H2S is modulated by sulfide quinone oxidoreductase, which commits sulfide to oxidative catabolism. Since induced hypometabolism can be beneficial in disease settings that involve insufficient or interrupted blood flow, our study has important implications for attenuating reperfusion-induced ischemic injury and/or prolonging the shelf life of biologics like platelets.


Subject(s)
Hydrogen Sulfide , Reperfusion Injury , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Sulfides , Oxidation-Reduction , Mammals/metabolism
14.
Proc Natl Acad Sci U S A ; 121(11): e2314199121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38451940

ABSTRACT

Proton-powered c-ring rotation in mitochondrial ATP synthase is crucial to convert the transmembrane protonmotive force into torque to drive the synthesis of adenosine triphosphate (ATP). Capitalizing on recent cryo-EM structures, we aim at a structural and energetic understanding of how functional directional rotation is achieved. We performed multi-microsecond atomistic simulations to determine the free energy profiles along the c-ring rotation angle before and after the arrival of a new proton. Our results reveal that rotation proceeds by dynamic sliding of the ring over the a-subunit surface, during which interactions with conserved polar residues stabilize distinct intermediates. Ordered water chains line up for a Grotthuss-type proton transfer in one of these intermediates. After proton transfer, a high barrier prevents backward rotation and an overall drop in free energy favors forward rotation, ensuring the directionality of c-ring rotation required for the thermodynamically disfavored ATP synthesis. The essential arginine of the a-subunit stabilizes the rotated configuration through a salt bridge with the c-ring. Overall, we describe a complete mechanism for the rotation step of the ATP synthase rotor, thereby illuminating a process critical to all life at atomic resolution.


Subject(s)
Mitochondrial Proton-Translocating ATPases , Protons , Mitochondrial Proton-Translocating ATPases/metabolism , Protein Conformation , Adenosine Triphosphate , Rotation , Proton-Translocating ATPases/metabolism
15.
EMBO J ; 41(12): e109049, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35319107

ABSTRACT

Cellular metabolism must adapt to changing demands to enable homeostasis. During immune responses or cancer metastasis, cells leading migration into challenging environments require an energy boost, but what controls this capacity is unclear. Here, we study a previously uncharacterized nuclear protein, Atossa (encoded by CG9005), which supports macrophage invasion into the germband of Drosophila by controlling cellular metabolism. First, nuclear Atossa increases mRNA levels of Porthos, a DEAD-box protein, and of two metabolic enzymes, lysine-α-ketoglutarate reductase (LKR/SDH) and NADPH glyoxylate reductase (GR/HPR), thus enhancing mitochondrial bioenergetics. Then Porthos supports ribosome assembly and thereby raises the translational efficiency of a subset of mRNAs, including those affecting mitochondrial functions, the electron transport chain, and metabolism. Mitochondrial respiration measurements, metabolomics, and live imaging indicate that Atossa and Porthos power up OxPhos and energy production to promote the forging of a path into tissues by leading macrophages. Since many crucial physiological responses require increases in mitochondrial energy output, this previously undescribed genetic program may modulate a wide range of cellular behaviors.


Subject(s)
Drosophila , Saccharopine Dehydrogenases , Animals , Drosophila/metabolism , Energy Metabolism , Macrophages/metabolism , Mitochondria/metabolism , RNA, Messenger/metabolism , Saccharopine Dehydrogenases/genetics , Saccharopine Dehydrogenases/metabolism
16.
Proc Natl Acad Sci U S A ; 120(31): e2306046120, 2023 08.
Article in English | MEDLINE | ID: mdl-37487099

ABSTRACT

The electron-conducting circuitry of life represents an as-yet untapped resource of exquisite, nanoscale biomolecular engineering. Here, we report the characterization and structure of a de novo diheme "maquette" protein, 4D2, which we subsequently use to create an expanded, modular platform for heme protein design. A well-folded monoheme variant was created by computational redesign, which was then utilized for the experimental validation of continuum electrostatic redox potential calculations. This demonstrates how fundamental biophysical properties can be predicted and fine-tuned. 4D2 was then extended into a tetraheme helical bundle, representing a 7 nm molecular wire. Despite a molecular weight of only 24 kDa, electron cryomicroscopy illustrated a remarkable level of detail, indicating the positioning of the secondary structure and the heme cofactors. This robust, expressible, highly thermostable and readily designable modular platform presents a valuable resource for redox protein design and the future construction of artificial electron-conducting circuitry.


Subject(s)
Hemeproteins , Biophysics , Cryoelectron Microscopy , Electrons , Oxidation-Reduction
17.
Proc Natl Acad Sci U S A ; 120(9): e2216810120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36812201

ABSTRACT

Mitochondria provide essential metabolites and adenosine triphosphate (ATP) for the regulation of energy homeostasis. For instance, liver mitochondria are a vital source of gluconeogenic precursors under a fasted state. However, the regulatory mechanisms at the level of mitochondrial membrane transport are not fully understood. Here, we report that a liver-specific mitochondrial inner-membrane carrier SLC25A47 is required for hepatic gluconeogenesis and energy homeostasis. Genome-wide association studies found significant associations between SLC25A47 and fasting glucose, HbA1c, and cholesterol levels in humans. In mice, we demonstrated that liver-specific depletion of SLC25A47 impaired hepatic gluconeogenesis selectively from lactate, while significantly enhancing whole-body energy expenditure and the hepatic expression of FGF21. These metabolic changes were not a consequence of general liver dysfunction because acute SLC25A47 depletion in adult mice was sufficient to enhance hepatic FGF21 production, pyruvate tolerance, and insulin tolerance independent of liver damage and mitochondrial dysfunction. Mechanistically, SLC25A47 depletion leads to impaired hepatic pyruvate flux and malate accumulation in the mitochondria, thereby restricting hepatic gluconeogenesis. Together, the present study identified a crucial node in the liver mitochondria that regulates fasting-induced gluconeogenesis and energy homeostasis.


Subject(s)
Genome-Wide Association Study , Gluconeogenesis , Humans , Mice , Animals , Gluconeogenesis/physiology , Glucose/metabolism , Liver/metabolism , Energy Metabolism/physiology , Pyruvates/metabolism
18.
Proc Natl Acad Sci U S A ; 120(8): e2215650120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36780529

ABSTRACT

F1-ATPase is a motor protein that couples the rotation of its rotary [Formula: see text] subunit with ATP synthesis or hydrolysis. Single-molecule experiments indicate that nucleotide binding and release events occur almost simultaneously during the synthesis cycle, allowing the energy gain due to spontaneous binding of ADP to one catalytic [Formula: see text] subunit to be directly harnessed for driving the release of ATP from another rather than being dissipated as heat. Here, we examine the unknown mechanism of this coupling that is critical for an exceptionally high mechanochemical efficiency of F1-ATPase by means of all-atom free-energy simulations. We find that nondissipative and kinetically fast progression of the motor in the synthesis direction requires a concerted conformational change involving the closure of the ADP-binding [Formula: see text] subunit followed by the gradual opening of the ATP-releasing [Formula: see text] subunit over the course of the 30 to 40° rotary substep of the [Formula: see text] subunit. This rotary substep, preceding the ATP-dependent metastable state, allows for the recovery of a large portion of the ADP binding energy in the conformation of ATP-bound [Formula: see text] that gradually adopts the low-affinity conformation, captured also by the recent cryo-EM structure of this elusive state. The release of ATP from this nearly open conformation leads to its further opening, which enables the progression of the motor to the next catalytic metastable state. Our simulations explain this energy conversion mechanism in terms of intersubunit and ligand-protein interactions.


Subject(s)
Adenosine Triphosphate , Proton-Translocating ATPases , Proton-Translocating ATPases/metabolism , Catalysis , Protein Conformation , Thermodynamics , Adenosine Triphosphate/metabolism , Hydrolysis , Kinetics
19.
Proc Natl Acad Sci U S A ; 120(52): e2306160120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38109545

ABSTRACT

Epulopiscium spp. are the largest known heterotrophic bacteria; a large cigar-shaped individual is a million times the volume of Escherichia coli. To better understand the metabolic potential and relationship of Epulopiscium sp. type B with its host Naso tonganus, we generated a high-quality draft genome from a population of cells taken from a single fish. We propose the name Candidatus Epulopiscium viviparus to describe populations of this best-characterized Epulopiscium species. Metabolic reconstruction reveals more than 5% of the genome codes for carbohydrate active enzymes, which likely degrade recalcitrant host-diet algal polysaccharides into substrates that may be fermented to acetate, the most abundant short-chain fatty acid in the intestinal tract. Moreover, transcriptome analyses and the concentration of sodium ions in the host intestinal tract suggest that the use of a sodium motive force (SMF) to drive ATP synthesis and flagellar rotation is integral to symbiont metabolism and cellular biology. In natural populations, genes encoding both F-type and V-type ATPases and SMF generation via oxaloacetate decarboxylation are among the most highly expressed, suggesting that ATPases synthesize ATP and balance ion concentrations across the cell membrane. High expression of these and other integral membrane proteins may allow for the growth of its extensive intracellular membrane system. Further, complementary metabolism between microbe and host is implied with the potential provision of nitrogen and B vitamins to reinforce this nutritional symbiosis. The few features shared by all bacterial behemoths include extreme polyploidy, polyphosphate synthesis, and thus far, they have all resisted cultivation in the lab.


Subject(s)
Sodium , Vacuolar Proton-Translocating ATPases , Animals , Sodium/metabolism , Bacteria/metabolism , Clostridiales/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/metabolism
20.
Proc Natl Acad Sci U S A ; 120(34): e2210924120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37579147

ABSTRACT

The origin and early evolution of life is generally studied under two different paradigms: bottom up and top down. Prebiotic chemistry and early Earth geochemistry allow researchers to explore possible origin of life scenarios. But for these "bottom-up" approaches, even successful experiments only amount to a proof of principle. On the other hand, "top-down" research on early evolutionary history is able to provide a historical account about ancient organisms, but is unable to investigate stages that occurred during and just after the origin of life. Here, we consider ancient electron transport chains (ETCs) as a potential bridge between early evolutionary history and a protocellular stage that preceded it. Current phylogenetic evidence suggests that ancestors of several extant ETC components were present at least as late as the last universal common ancestor of life. In addition, recent experiments have shown that some aspects of modern ETCs can be replicated by minerals, protocells, or organic cofactors in the absence of biological proteins. Here, we discuss the diversity of ETCs and other forms of chemiosmotic energy conservation, describe current work on the early evolution of membrane bioenergetics, and advocate for several lines of research to enhance this understanding by pairing top-down and bottom-up approaches.


Subject(s)
Biochemical Phenomena , Phylogeny , Electron Transport , Proteins/chemistry , Energy Metabolism , Origin of Life , Biological Evolution , Evolution, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL