Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.625
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 185(20): 3720-3738.e13, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36103894

RESUMEN

Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals.


Asunto(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animales , Mycobacterium tuberculosis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra
2.
Cell ; 184(23): 5824-5837.e15, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34672953

RESUMEN

The human mitochondrial genome encodes thirteen core subunits of the oxidative phosphorylation system, and defects in mitochondrial gene expression lead to severe neuromuscular disorders. However, the mechanisms of mitochondrial gene expression remain poorly understood due to a lack of experimental approaches to analyze these processes. Here, we present an in vitro system to silence translation in purified mitochondria. In vitro import of chemically synthesized precursor-morpholino hybrids allows us to target translation of individual mitochondrial mRNAs. By applying this approach, we conclude that the bicistronic, overlapping ATP8/ATP6 transcript is translated through a single ribosome/mRNA engagement. We show that recruitment of COX1 assembly factors to translating ribosomes depends on nascent chain formation. By defining mRNA-specific interactomes for COX1 and COX2, we reveal an unexpected function of the cytosolic oncofetal IGF2BP1, an RNA-binding protein, in mitochondrial translation. Our data provide insight into mitochondrial translation and innovative strategies to investigate mitochondrial gene expression.


Asunto(s)
Regulación de la Expresión Génica , Silenciador del Gen , Genes Mitocondriales , Transporte de Electrón , Complejo IV de Transporte de Electrones/genética , Células HEK293 , Humanos , Proteínas Mitocondriales/metabolismo , Oligonucleótidos/química , Fosforilación Oxidativa , Biosíntesis de Proteínas , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mitocondrial/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Saccharomyces cerevisiae/metabolismo
3.
Cell ; 184(21): 5375-5390.e16, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34562363

RESUMEN

Although oxidative phosphorylation is best known for producing ATP, it also yields reactive oxygen species (ROS) as invariant byproducts. Depletion of ROS below their physiological levels, a phenomenon known as reductive stress, impedes cellular signaling and has been linked to cancer, diabetes, and cardiomyopathy. Cells alleviate reductive stress by ubiquitylating and degrading the mitochondrial gatekeeper FNIP1, yet it is unknown how the responsible E3 ligase CUL2FEM1B can bind its target based on redox state and how this is adjusted to changing cellular environments. Here, we show that CUL2FEM1B relies on zinc as a molecular glue to selectively recruit reduced FNIP1 during reductive stress. FNIP1 ubiquitylation is gated by pseudosubstrate inhibitors of the BEX family, which prevent premature FNIP1 degradation to protect cells from unwarranted ROS accumulation. FEM1B gain-of-function mutation and BEX deletion elicit similar developmental syndromes, showing that the zinc-dependent reductive stress response must be tightly regulated to maintain cellular and organismal homeostasis.


Asunto(s)
Estrés Fisiológico , Aminoácidos/química , Animales , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Femenino , Humanos , Iones , Ratones , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico/efectos de los fármacos , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , Complejos de Ubiquitina-Proteína Ligasa/química , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitinación/efectos de los fármacos , Zinc/farmacología
4.
Cell ; 180(6): 1160-1177.e20, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32160526

RESUMEN

Selective autophagy of organelles is critical for cellular differentiation, homeostasis, and organismal health. Autophagy of the ER (ER-phagy) is implicated in human neuropathy but is poorly understood beyond a few autophagosomal receptors and remodelers. By using an ER-phagy reporter and genome-wide CRISPRi screening, we identified 200 high-confidence human ER-phagy factors. Two pathways were unexpectedly required for ER-phagy. First, reduced mitochondrial metabolism represses ER-phagy, which is opposite of general autophagy and is independent of AMPK. Second, ER-localized UFMylation is required for ER-phagy to repress the unfolded protein response via IRE1α. The UFL1 ligase is brought to the ER surface by DDRGK1 to UFMylate RPN1 and RPL26 and preferentially targets ER sheets for degradation, analogous to PINK1-Parkin regulation during mitophagy. Our data provide insight into the cellular logic of ER-phagy, reveal parallels between organelle autophagies, and provide an entry point to the relatively unexplored process of degrading the ER network.


Asunto(s)
Autofagia/fisiología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Autofagia/genética , Estrés del Retículo Endoplásmico/fisiología , Endorribonucleasas/metabolismo , Estudio de Asociación del Genoma Completo/métodos , Células HCT116 , Células HEK293 , Células HeLa , Homeostasis , Humanos , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Respuesta de Proteína Desplegada/fisiología
5.
Cell ; 182(5): 1170-1185.e9, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32795412

RESUMEN

Loss of the gene (Fmr1) encoding Fragile X mental retardation protein (FMRP) causes increased mRNA translation and aberrant synaptic development. We find neurons of the Fmr1-/y mouse have a mitochondrial inner membrane leak contributing to a "leak metabolism." In human Fragile X syndrome (FXS) fibroblasts and in Fmr1-/y mouse neurons, closure of the ATP synthase leak channel by mild depletion of its c-subunit or pharmacological inhibition normalizes stimulus-induced and constitutive mRNA translation rate, decreases lactate and key glycolytic and tricarboxylic acid (TCA) cycle enzyme levels, and triggers synapse maturation. FMRP regulates leak closure in wild-type (WT), but not FX synapses, by stimulus-dependent ATP synthase ß subunit translation; this increases the ratio of ATP synthase enzyme to its c-subunit, enhancing ATP production efficiency and synaptic growth. In contrast, in FXS, inability to close developmental c-subunit leak prevents stimulus-dependent synaptic maturation. Therefore, ATP synthase c-subunit leak closure encourages development and attenuates autistic behaviors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Subunidades de Proteína/metabolismo , Animales , Línea Celular , Ciclo del Ácido Cítrico/fisiología , Fibroblastos/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Células HEK293 , Humanos , Ratones , Neuronas/metabolismo , ARN Mensajero , Sinapsis/metabolismo
6.
Cell ; 174(4): 982-998.e20, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29909982

RESUMEN

The diversity of cell types and regulatory states in the brain, and how these change during aging, remains largely unknown. We present a single-cell transcriptome atlas of the entire adult Drosophila melanogaster brain sampled across its lifespan. Cell clustering identified 87 initial cell clusters that are further subclustered and validated by targeted cell-sorting. Our data show high granularity and identify a wide range of cell types. Gene network analyses using SCENIC revealed regulatory heterogeneity linked to energy consumption. During aging, RNA content declines exponentially without affecting neuronal identity in old brains. This single-cell brain atlas covers nearly all cells in the normal brain and provides the tools to study cellular diversity alongside other Drosophila and mammalian single-cell datasets in our unique single-cell analysis platform: SCope (http://scope.aertslab.org). These results, together with SCope, allow comprehensive exploration of all transcriptional states of an entire aging brain.


Asunto(s)
Envejecimiento , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Redes Reguladoras de Genes , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Drosophila melanogaster/fisiología , Femenino , Perfilación de la Expresión Génica , Masculino
7.
Immunity ; 56(3): 516-530.e9, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36738738

RESUMEN

In vitro studies have associated oxidative phosphorylation (OXPHOS) with anti-inflammatory macrophages, whereas pro-inflammatory macrophages rely on glycolysis. However, the metabolic needs of macrophages in tissues (TMFs) to fulfill their homeostatic activities are incompletely understood. Here, we identified OXPHOS as the highest discriminating process among TMFs from different organs in homeostasis by analysis of RNA-seq data in both humans and mice. Impairing OXPHOS in TMFs via Tfam deletion differentially affected TMF populations. Tfam deletion resulted in reduction of alveolar macrophages (AMs) due to impaired lipid-handling capacity, leading to increased cholesterol content and cellular stress, causing cell-cycle arrest in vivo. In obesity, Tfam depletion selectively ablated pro-inflammatory lipid-handling white adipose tissue macrophages (WAT-MFs), thus preventing insulin resistance and hepatosteatosis. Hence, OXPHOS, rather than glycolysis, distinguishes TMF populations and is critical for the maintenance of TMFs with a high lipid-handling activity, including pro-inflammatory WAT-MFs. This could provide a selective therapeutic targeting tool.


Asunto(s)
Inflamación , Fosforilación Oxidativa , Humanos , Ratones , Animales , Inflamación/metabolismo , Macrófagos/metabolismo , Homeostasis , Lípidos , Tejido Adiposo/metabolismo
8.
Cell ; 170(6): 1247-1257.e12, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28844695

RESUMEN

The respiratory megacomplex represents the highest-order assembly of respiratory chain complexes, and it allows mitochondria to respond to energy-requiring conditions. To understand its architecture, we examined the human respiratory chain megacomplex-I2III2IV2 (MCI2III2IV2) with 140 subunits and a subset of associated cofactors using cryo-electron microscopy. The MCI2III2IV2 forms a circular structure with the dimeric CIII located in the center, where it is surrounded by two copies each of CI and CIV. Two cytochrome c (Cyt.c) molecules are positioned to accept electrons on the surface of the c1 state CIII dimer. Analyses indicate that CII could insert into the gaps between CI and CIV to form a closed ring, which we termed the electron transport chain supercomplex. The structure not only reveals the precise assignment of individual subunits of human CI and CIII, but also enables future in-depth analysis of the electron transport chain as a whole.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/química , Complejos Multienzimáticos/química , Microscopía por Crioelectrón , Proteínas del Complejo de Cadena de Transporte de Electrón/aislamiento & purificación , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/aislamiento & purificación , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/aislamiento & purificación , Complejo II de Transporte de Electrones/metabolismo , Humanos , Mitocondrias/química , Mitocondrias/metabolismo , Modelos Moleculares , Complejos Multienzimáticos/aislamiento & purificación , Complejos Multienzimáticos/metabolismo
9.
Mol Cell ; 84(8): 1541-1555.e11, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38503286

RESUMEN

Oxidative phosphorylation (OXPHOS) complexes, encoded by both mitochondrial and nuclear DNA, are essential producers of cellular ATP, but how nuclear and mitochondrial gene expression steps are coordinated to achieve balanced OXPHOS subunit biogenesis remains unresolved. Here, we present a parallel quantitative analysis of the human nuclear and mitochondrial messenger RNA (mt-mRNA) life cycles, including transcript production, processing, ribosome association, and degradation. The kinetic rates of nearly every stage of gene expression differed starkly across compartments. Compared with nuclear mRNAs, mt-mRNAs were produced 1,100-fold more, degraded 7-fold faster, and accumulated to 160-fold higher levels. Quantitative modeling and depletion of mitochondrial factors LRPPRC and FASTKD5 identified critical points of mitochondrial regulatory control, revealing that the mitonuclear expression disparities intrinsically arise from the highly polycistronic nature of human mitochondrial pre-mRNA. We propose that resolving these differences requires a 100-fold slower mitochondrial translation rate, illuminating the mitoribosome as a nexus of mitonuclear co-regulation.


Asunto(s)
Mitocondrias , Ribosomas Mitocondriales , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Ribosomas Mitocondriales/metabolismo , Biosíntesis de Proteínas , Fosforilación Oxidativa , Proteínas Mitocondriales/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo
10.
Cell ; 167(6): 1598-1609.e10, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27912063

RESUMEN

The mammalian respiratory chain complexes assemble into supercomplexes (SCs) and reside in the inner mitochondrial membrane to transfer electrons and establish the proton gradient for complex V to synthesize ATP. The precise arrangement of SCs is largely unknown. Here, we report a 4.0-Å cryo-electron microscopy (cryo-EM) structure of the major SC in porcine heart, the 1.7-MDa SCI1III2IV1. The complex III (CIII) dimer and complex IV (CIV) bind at the same side of the L-shaped complex I (CI). Several accessory or supernumerary subunits of CI, such as NDUFA11, NDUFB4, NDUFB8, and NDUFB9, directly contribute to the oligomerization of CI, CIII, and CIV. COX7C and COX7A of CIV attach CIV to the concave surface formed by CIII and the distal end of membrane arm of CI. The structure suggests a possible mechanism by which electrons are transferred from NADH to cytochrome c and provides a platform for future functional dissection of respiration.


Asunto(s)
Transporte de Electrón , Mitocondrias Cardíacas/química , Membranas Mitocondriales/química , Animales , Microscopía por Crioelectrón , Modelos Moleculares , Complejos Multienzimáticos/química , Bombas de Protones/química , Sus scrofa
11.
Mol Cell ; 83(21): 3904-3920.e7, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37879334

RESUMEN

Lactate has long been considered a cellular waste product. However, we found that as extracellular lactate accumulates, it also enters the mitochondrial matrix and stimulates mitochondrial electron transport chain (ETC) activity. The resulting increase in mitochondrial ATP synthesis suppresses glycolysis and increases the utilization of pyruvate and/or alternative respiratory substrates. The ability of lactate to increase oxidative phosphorylation does not depend on its metabolism. Both L- and D-lactate are effective at enhancing ETC activity and suppressing glycolysis. Furthermore, the selective induction of mitochondrial oxidative phosphorylation by unmetabolized D-lactate reversibly suppressed aerobic glycolysis in both cancer cell lines and proliferating primary cells in an ATP-dependent manner and enabled cell growth on respiratory-dependent bioenergetic substrates. In primary T cells, D-lactate enhanced cell proliferation and effector function. Together, these findings demonstrate that lactate is a critical regulator of the ability of mitochondrial oxidative phosphorylation to suppress glucose fermentation.


Asunto(s)
Metabolismo Energético , Ácido Láctico , Ácido Láctico/metabolismo , Transporte de Electrón , Fosforilación Oxidativa , Glucólisis/fisiología , Adenosina Trifosfato/metabolismo
12.
Mol Cell ; 82(18): 3321-3332, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35961309

RESUMEN

Mitochondrial energetics and respiration have emerged as important factors in how cancer cells respond to or evade apoptotic signals. The study of the functional connection between these two processes may provide insight into following questions old and new: how might we target respiration or downstream signaling pathways to amplify apoptotic stress in the context of cancer therapy? Why are respiration and apoptotic regulation housed in the same organelle? Here, we briefly review mitochondrial respiration and apoptosis and then focus on how the intersection of these two processes is regulated by cytoplasmic signaling pathways such as the integrated stress response.


Asunto(s)
Mitocondrias , Neoplasias , Apoptosis , Humanos , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Estrés Oxidativo , Respiración , Transducción de Señal
13.
Physiol Rev ; 101(4): 1561-1607, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33733879

RESUMEN

The design of the energy metabolism system in striated muscle remains a major area of investigation. Here, we review our current understanding and emerging hypotheses regarding the metabolic support of muscle contraction. Maintenance of ATP free energy, so called energy homeostasis, via mitochondrial oxidative phosphorylation is critical to sustained contractile activity, and this major design criterion is the focus of this review. Cell volume invested in mitochondria reduces the space available for generating contractile force, and this spatial balance between mitochondria acontractile elements to meet the varying sustained power demands across muscle types is another important design criterion. This is accomplished with remarkably similar mass-specific mitochondrial protein composition across muscle types, implying that it is the organization of mitochondria within the muscle cell that is critical to supporting sustained muscle function. Beyond the production of ATP, ubiquitous distribution of ATPases throughout the muscle requires rapid distribution of potential energy across these large cells. Distribution of potential energy has long been thought to occur primarily through facilitated metabolite diffusion, but recent analysis has questioned the importance of this process under normal physiological conditions. Recent structural and functional studies have supported the hypothesis that the mitochondrial reticulum provides a rapid energy distribution system via the conduction of the mitochondrial membrane potential to maintain metabolic homeostasis during contractile activity. We extensively review this aspect of the energy metabolism design contrasting it with metabolite diffusion models and how mitochondrial structure can play a role in the delivery of energy in the striated muscle.


Asunto(s)
Metabolismo Energético/fisiología , Músculo Estriado/metabolismo , Animales , Humanos , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/fisiología , Células Musculares/metabolismo
14.
EMBO J ; 43(14): 2979-3008, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38839991

RESUMEN

Lipid-protein interactions play a multitude of essential roles in membrane homeostasis. Mitochondrial membranes have a unique lipid-protein environment that ensures bioenergetic efficiency. Cardiolipin (CL), the signature mitochondrial lipid, plays multiple roles in promoting oxidative phosphorylation (OXPHOS). In the inner mitochondrial membrane, the ADP/ATP carrier (AAC in yeast; adenine nucleotide translocator, ANT in mammals) exchanges ADP and ATP, enabling OXPHOS. AAC/ANT contains three tightly bound CLs, and these interactions are evolutionarily conserved. Here, we investigated the role of these buried CLs in AAC/ANT using a combination of biochemical approaches, native mass spectrometry, and molecular dynamics simulations. We introduced negatively charged mutations into each CL-binding site of yeast Aac2 and established experimentally that the mutations disrupted the CL interactions. While all mutations destabilized Aac2 tertiary structure, transport activity was impaired in a binding site-specific manner. Additionally, we determined that a disease-associated missense mutation in one CL-binding site in human ANT1 compromised its structure and transport activity, resulting in OXPHOS defects. Our findings highlight the conserved significance of CL in AAC/ANT structure and function, directly tied to specific lipid-protein interactions.


Asunto(s)
Cardiolipinas , Translocasas Mitocondriales de ADP y ATP , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Cardiolipinas/metabolismo , Sitios de Unión , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Humanos , Translocasas Mitocondriales de ADP y ATP/metabolismo , Translocasas Mitocondriales de ADP y ATP/genética , Translocasas Mitocondriales de ADP y ATP/química , Fosforilación Oxidativa , Translocador 1 del Nucleótido Adenina/metabolismo , Translocador 1 del Nucleótido Adenina/genética , Simulación de Dinámica Molecular , Unión Proteica , Mitocondrias/metabolismo , Mitocondrias/genética , Membranas Mitocondriales/metabolismo , Mutación , Mutación Missense
15.
Trends Biochem Sci ; 48(5): 463-476, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36702698

RESUMEN

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.


Asunto(s)
Enfermedades Mitocondriales , Ubiquinona , Humanos , Ubiquinona/metabolismo , Enfermedades Mitocondriales/metabolismo , Oxidación-Reducción , Ataxia/metabolismo , Lípidos
16.
Plant Cell ; 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39179507

RESUMEN

EARLY NODULIN 93 (ENOD93) has been genetically associated with biological nitrogen fixation in legumes and nitrogen use efficiency in cereals, but its precise function is unknown. We show that hidden Markov models define ENOD93 as a homolog of the N-terminal domain of RESPIRATORY SUPERCOMPLEX FACTOR 2 (RCF2). RCF2 regulates cytochrome oxidase (CIV), influencing the generation of a mitochondrial proton motive force in yeast (Saccharomyces cerevisiae). Knockout of ENOD93 in Arabidopsis (Arabidopsis thaliana) causes a short root phenotype and early flowering. ENOD93 is associated with a protein complex the size of CIV in mitochondria, but neither CIV abundance nor its activity changed in ruptured organelles of enod93. However, a progressive loss of ADP-dependent respiration rate was observed in intact enod93 mitochondria, which could be recovered in complemented lines. Mitochondrial membrane potential was higher in enod93 in a CIV-dependent manner, but ATP synthesis and ADP depletion rates progressively decreased. The respiration rate of whole enod93 seedlings was elevated, and root ADP content was nearly double that in wild type without a change in ATP content. We propose that ENOD93 and HYPOXIA-INDUCED GENE DOMAIN 2 (HIGD2) are the functional equivalent of yeast RCF2 but have remained undiscovered in many eukaryotic lineages because they are encoded by two distinct genes.

17.
Mol Cell ; 75(6): 1131-1146.e6, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31492636

RESUMEN

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.


Asunto(s)
Complejo III de Transporte de Electrones/química , Complejo I de Transporte de Electrón/química , Mitocondrias Cardíacas/enzimología , Animales , Cristalografía por Rayos X , Estructura Cuaternaria de Proteína , Ovinos
18.
Hum Mol Genet ; 33(R1): R34-R41, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38779776

RESUMEN

In human cells, the nuclear and mitochondrial genomes engage in a complex interplay to produce dual-encoded oxidative phosphorylation (OXPHOS) complexes. The coordination of these dynamic gene expression processes is essential for producing matched amounts of OXPHOS protein subunits. This review focuses on our current understanding of the mitochondrial central dogma rates, highlighting the striking differences in gene expression rates between mitochondrial and nuclear genes. We synthesize a coherent model of mitochondrial gene expression kinetics, highlighting the emerging principles and emphasizing where more precise measurements would be beneficial. Such an understanding is pivotal for grasping the unique aspects of mitochondrial function and its role in cellular energetics, and it has profound implications for aging, metabolic disorders, and neurodegenerative diseases.


Asunto(s)
Mitocondrias , Fosforilación Oxidativa , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Regulación de la Expresión Génica , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Genoma Mitocondrial , Metabolismo Energético/genética , Núcleo Celular/metabolismo , Núcleo Celular/genética , Envejecimiento/genética , Envejecimiento/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo
19.
Hum Mol Genet ; 33(R1): R47-R52, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38779773

RESUMEN

The mitochondrial oxidative phosphorylation (OXPHOS) system produces the majority of energy required by cells. Given the mitochondrion's endosymbiotic origin, the OXPHOS machinery is still under dual genetic control where most OXPHOS subunits are encoded by the nuclear DNA and imported into mitochondria, while a small subset is encoded on the mitochondrion's own genome, the mitochondrial DNA (mtDNA). The nuclear and mtDNA encoded subunits must be expressed and assembled in a highly orchestrated fashion to form a functional OXPHOS system and meanwhile prevent the generation of any harmful assembly intermediates. While several mechanisms have evolved in eukaryotes to achieve such a coordinated expression, this review will focus on how the translation of mtDNA encoded OXPHOS subunits is tailored to OXPHOS assembly.


Asunto(s)
ADN Mitocondrial , Mitocondrias , Fosforilación Oxidativa , Biosíntesis de Proteínas , Mitocondrias/metabolismo , Mitocondrias/genética , Humanos , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Animales
20.
EMBO J ; 41(12): e109049, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35319107

RESUMEN

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.


Asunto(s)
Drosophila , Sacaropina Deshidrogenasas , Animales , Drosophila/metabolismo , Metabolismo Energético , Macrófagos/metabolismo , Mitocondrias/metabolismo , ARN Mensajero/metabolismo , Sacaropina Deshidrogenasas/genética , Sacaropina Deshidrogenasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA