Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Cell ; 186(11): 2361-2379.e25, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37192619

ABSTRACT

Multiple anticancer drugs have been proposed to cause cell death, in part, by increasing the steady-state levels of cellular reactive oxygen species (ROS). However, for most of these drugs, exactly how the resultant ROS function and are sensed is poorly understood. It remains unclear which proteins the ROS modify and their roles in drug sensitivity/resistance. To answer these questions, we examined 11 anticancer drugs with an integrated proteogenomic approach identifying not only many unique targets but also shared ones-including ribosomal components, suggesting common mechanisms by which drugs regulate translation. We focus on CHK1 that we find is a nuclear H2O2 sensor that launches a cellular program to dampen ROS. CHK1 phosphorylates the mitochondrial DNA-binding protein SSBP1 to prevent its mitochondrial localization, which in turn decreases nuclear H2O2. Our results reveal a druggable nucleus-to-mitochondria ROS-sensing pathway-required to resolve nuclear H2O2 accumulation and mediate resistance to platinum-based agents in ovarian cancers.


Subject(s)
Antineoplastic Agents , Reactive Oxygen Species , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Cell Nucleus/metabolism , Humans
2.
Mol Cell ; 84(2): 345-358.e5, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38199007

ABSTRACT

Cellular proteostasis requires transport of polypeptides across membranes. Although defective transport processes trigger cytosolic rescue and quality control mechanisms that clear translocases and membranes from unproductive cargo, proteins that are synthesized within mitochondria are not accessible to these mechanisms. Mitochondrial-encoded proteins are inserted cotranslationally into the inner membrane by the conserved insertase OXA1L. Here, we identify TMEM126A as a OXA1L-interacting protein. TMEM126A associates with mitochondrial ribosomes and translation products. Loss of TMEM126A leads to the destabilization of mitochondrial translation products, triggering an inner membrane quality control process, in which newly synthesized proteins are degraded by the mitochondrial iAAA protease. Our data reveal that TMEM126A cooperates with OXA1L in protein insertion into the membrane. Upon loss of TMEM126A, the cargo-blocked OXA1L insertase complexes undergo proteolytic clearance by the iAAA protease machinery together with its cargo.


Subject(s)
Mitochondria , Mitochondrial Membranes , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Peptide Hydrolases/metabolism
3.
Mol Cell ; 84(8): 1541-1555.e11, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38503286

ABSTRACT

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.


Subject(s)
Mitochondria , Mitochondrial Ribosomes , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Ribosomes/metabolism , Protein Biosynthesis , Oxidative Phosphorylation , Mitochondrial Proteins/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism
4.
Annu Rev Biochem ; 85: 103-32, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27023846

ABSTRACT

Mitochondrial ribosomes (mitoribosomes) perform protein synthesis inside mitochondria, the organelles responsible for energy conversion and adenosine triphosphate production in eukaryotic cells. Throughout evolution, mitoribosomes have become functionally specialized for synthesizing mitochondrial membrane proteins, and this has been accompanied by large changes to their structure and composition. We review recent high-resolution structural data that have provided unprecedented insight into the structure and function of mitoribosomes in mammals and fungi.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Ribosomes/ultrastructure , Protein Biosynthesis , Ribosome Subunits/ultrastructure , Animals , Anti-Bacterial Agents/pharmacology , Biological Evolution , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , DNA, Mitochondrial/metabolism , Mammals , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/chemistry , Mitochondrial Ribosomes/metabolism , Models, Molecular , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribosome Subunits/chemistry , Ribosome Subunits/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
5.
Cell ; 167(2): 471-483.e10, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693358

ABSTRACT

Mitochondrial ribosomes translate membrane integral core subunits of the oxidative phosphorylation system encoded by mtDNA. These translation products associate with nuclear-encoded, imported proteins to form enzyme complexes that produce ATP. Here, we show that human mitochondrial ribosomes display translational plasticity to cope with the supply of imported nuclear-encoded subunits. Ribosomes expressing mitochondrial-encoded COX1 mRNA selectively engage with cytochrome c oxidase assembly factors in the inner membrane. Assembly defects of the cytochrome c oxidase arrest mitochondrial translation in a ribosome nascent chain complex with a partially membrane-inserted COX1 translation product. This complex represents a primed state of the translation product that can be retrieved for assembly. These findings establish a mammalian translational plasticity pathway in mitochondria that enables adaptation of mitochondrial protein synthesis to the influx of nuclear-encoded subunits.


Subject(s)
Cyclooxygenase 1/metabolism , Electron Transport Complex IV/metabolism , Membrane Proteins/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Active Transport, Cell Nucleus , Cell Line, Tumor , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , DNA, Mitochondrial/genetics , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/genetics , HEK293 Cells , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Oxidative Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Mitochondrial , Ribosomes/metabolism
6.
Immunity ; 54(1): 68-83.e6, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33238133

ABSTRACT

While antibiotics are intended to specifically target bacteria, most are known to affect host cell physiology. In addition, some antibiotic classes are reported as immunosuppressive for reasons that remain unclear. Here, we show that Linezolid, a ribosomal-targeting antibiotic (RAbo), effectively blocked the course of a T cell-mediated autoimmune disease. Linezolid and other RAbos were strong inhibitors of T helper-17 cell effector function in vitro, showing that this effect was independent of their antibiotic activity. Perturbing mitochondrial translation in differentiating T cells, either with RAbos or through the inhibition of mitochondrial elongation factor G1 (mEF-G1) progressively compromised the integrity of the electron transport chain. Ultimately, this led to deficient oxidative phosphorylation, diminishing nicotinamide adenine dinucleotide concentrations and impairing cytokine production in differentiating T cells. In accordance, mice lacking mEF-G1 in T cells were protected from experimental autoimmune encephalomyelitis, demonstrating that this pathway is crucial in maintaining T cell function and pathogenicity.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Linezolid/therapeutic use , Mitochondria/metabolism , Peptides, Cyclic/therapeutic use , Ribosomes/metabolism , Th17 Cells/physiology , Animals , Autoimmunity/drug effects , Cell Differentiation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Targeted Therapy , Multiple Sclerosis/drug therapy , NAD/metabolism , Oxidative Phosphorylation , Peptide Elongation Factor G/genetics , Peptide Elongation Factor G/metabolism
7.
Hum Mol Genet ; 33(R1): R42-R46, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38779770

ABSTRACT

Mitochondrial translation is a complex process responsible for the synthesis of essential proteins involved in oxidative phosphorylation, a fundamental pathway for cellular energy production. Central to this process is the termination phase, where dedicated factors play a pivotal role in ensuring accurate and timely protein production. This review provides a comprehensive overview of the current understanding of translation termination in human mitochondria, emphasizing structural features and molecular functions of two mitochondrial termination factors mtRF1 and mtRF1a.


Subject(s)
Mitochondria , Mitochondrial Proteins , Peptide Chain Termination, Translational , Protein Biosynthesis , Humans , Mitochondria/metabolism , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Oxidative Phosphorylation , Peptide Termination Factors/metabolism , Peptide Termination Factors/genetics
8.
EMBO J ; 41(23): e111550, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36314841

ABSTRACT

Phosphoglycerate dehydrogenase (PHGDH) is a key serine biosynthesis enzyme whose aberrant expression promotes various types of tumors. Recently, PHGDH has been found to have some non-canonical functions beyond serine biosynthesis, but its specific mechanisms in tumorigenesis remain unclear. Here, we show that PHGDH localizes to the inner mitochondrial membrane and promotes the translation of mitochondrial DNA (mtDNA)-encoded proteins in liver cancer cells. Mechanistically, we demonstrate that mitochondrial PHGDH directly interacts with adenine nucleotide translocase 2 (ANT2) and then recruits mitochondrial elongation factor G2 (mtEFG2) to promote mitochondrial ribosome recycling efficiency, thereby promoting mtDNA-encoded protein expression and subsequent mitochondrial respiration. Moreover, we show that treatment with a mitochondrial translation inhibitor or depletion of mtEFG2 diminishes PHGDH-mediated tumor growth. Collectively, our findings uncover a previously unappreciated function of PHGDH in tumorigenesis acting via promotion of mitochondrial translation and bioenergetics.


Subject(s)
Liver Neoplasms , Phosphoglycerate Dehydrogenase , Humans , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Cell Line, Tumor , Serine , Liver Neoplasms/genetics , Carcinogenesis , DNA, Mitochondrial
9.
EMBO J ; 41(18): e109353, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35920020

ABSTRACT

Macrophage polarization is a process whereby macrophages acquire distinct effector states (M1 or M2) to carry out multiple and sometimes opposite functions. We show here that translational reprogramming occurs during macrophage polarization and that this relies on the Elongator complex subunit Elp3, an enzyme that modifies the wobble uridine base U34 in cytosolic tRNAs. Elp3 expression is downregulated by classical M1-activating signals in myeloid cells, where it limits the production of pro-inflammatory cytokines via FoxO1 phosphorylation, and attenuates experimental colitis in mice. In contrast, alternative M2-activating signals upregulate Elp3 expression through a PI3K- and STAT6-dependent signaling pathway. The metabolic reprogramming linked to M2 macrophage polarization relies on Elp3 and the translation of multiple candidates, including the mitochondrial ribosome large subunit proteins Mrpl3, Mrpl13, and Mrpl47. By promoting translation of its activator Ric8b in a codon-dependent manner, Elp3 also regulates mTORC2 activation. Elp3 expression in myeloid cells further promotes Wnt-driven tumor initiation in the intestine by maintaining a pool of tumor-associated macrophages exhibiting M2 features. Collectively, our data establish a functional link between tRNA modifications, mTORC2 activation, and macrophage polarization.


Subject(s)
Histone Acetyltransferases , Macrophage Activation , Signal Transduction , Animals , Codon/metabolism , Histone Acetyltransferases/genetics , Macrophage Activation/genetics , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice
10.
J Biol Chem ; 300(4): 107176, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499152

ABSTRACT

Mitochondrial translation depends on mRNA-specific activators. In Schizosaccharomyces pombe, DEAD-box protein Mrh5, pentatricopeptide repeat (PPR) protein Ppr4, Mtf2, and Sls1 form a stable complex (designated Mrh5C) required for translation of mitochondrial DNA (mtDNA)-encoded cox1 mRNA, the largest subunit of the cytochrome c oxidase complex. However, how Mrh5C is formed and what role Mrh5C plays in cox1 mRNA translation have not been reported. To address these questions, we investigated the role of individual Mrh5C subunits in the assembly and function of Mrh5C. Our results revealed that Mtf2 and Sls1 form a subcomplex that serves as a scaffold to bring Mrh5 and Ppr4 together. Mrh5C binds to the small subunit of the mitoribosome (mtSSU), but each subunit could not bind to the mtSSU independently. Importantly, Mrh5C is required for the association of cox1 mRNA with the mtSSU. Finally, we investigated the importance of the signature DEAD-box in Mrh5. We found that the DEAD-box of Mrh5 is required for the association of Mrh5C and cox1 mRNA with the mtSSU. Unexpectedly, this motif is also required for the interaction of Mrh5 with other Mrh5C subunits. Altogether, our results suggest that Mrh5 and Ppr4 cooperate in activating the translation of cox1 mRNA. Our results also suggest that Mrh5C activates the translation of cox1 mRNA by promoting the recruitment of cox1 mRNA to the mtSSU.


Subject(s)
Electron Transport Complex IV , Membrane Proteins , Mitochondrial Proteins , Protein Biosynthesis , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Membrane Proteins/metabolism
11.
Mol Cell ; 68(3): 540-551.e5, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100054

ABSTRACT

Mitochondrial function is controlled by two separate genomes. This feature makes mitochondria prone to proteotoxic stress when a stoichiometric imbalance occurs in the protein complexes that perform oxidative phosphorylation, which consist of both nuclear- and mitochondrial-encoded proteins. Such a proteotoxic stress is known to induce the mitochondrial unfolded protein response (UPRmt) in animals. It is unknown whether UPRmt occurs in plants. Here, we induced a mitonuclear protein imbalance in Arabidopsis through chemical or genetic interference. Mitochondrial proteotoxic stress activated a plant-specific UPRmt and impaired plant growth and development. The plant UPRmt pathway is triggered by a transient oxidative burst, activating MAPK and hormonal (involving ethylene and auxin) signaling, which are all geared to repair proteostasis. This also establishes phytohormones as bona fide plant mitokines. Our data ascertain that mitochondrial protein quality control pathways, such as the UPRmt, are conserved in plants and that hormone signaling is an essential mediator that regulates mitochondrial proteostasis.


Subject(s)
Arabidopsis/metabolism , Ethylenes/metabolism , Indoleacetic Acids/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Plant Growth Regulators/metabolism , Plant Proteins/metabolism , Plants, Genetically Modified/metabolism , Stress, Physiological , Unfolded Protein Response , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis/growth & development , Doxycycline/pharmacology , Gene Expression Regulation, Plant , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Plant Proteins/genetics , Plants, Genetically Modified/drug effects , Plants, Genetically Modified/genetics , Plants, Genetically Modified/growth & development , Protein Interaction Maps , Respiratory Burst , Signal Transduction , Time Factors , Unfolded Protein Response/drug effects
12.
EMBO Rep ; 23(2): e53081, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34866316

ABSTRACT

Mouse embryonic stem cells (mESCs) can self-renew indefinitely and maintain pluripotency. Inhibition of mechanistic target of rapamycin (mTOR) by the kinase inhibitor INK128 is known to induce paused pluripotency in mESCs cultured with traditional serum/LIF medium (SL), but the underlying mechanisms remain unclear. In this study, we demonstrate that mTOR complex 1 (mTORC1) but not complex 2 (mTORC2) mediates mTOR inhibition-induced paused pluripotency in cells grown in both SL and 2iL medium (GSK3 and MEK inhibitors and LIF). We also show that mTORC1 regulates self-renewal in both conditions mainly through eIF4F-mediated translation initiation that targets mRNAs of both cytosolic and mitochondrial ribosome subunits. Moreover, inhibition of mitochondrial translation is sufficient to induce paused pluripotency. Interestingly, eIF4F also regulates maintenance of pluripotency in an mTORC1-independent but MEK/ERK-dependent manner in SL, indicating that translation of pluripotency genes is controlled differently in SL and 2iL. Our study reveals a detailed picture of how mTOR governs self-renewal in mESCs and uncovers a context-dependent function of eIF4F in pluripotency regulation.


Subject(s)
Eukaryotic Initiation Factor-4F , Mechanistic Target of Rapamycin Complex 1 , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Eukaryotic Initiation Factor-4F/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2 , Mice
13.
Cell Mol Life Sci ; 80(12): 361, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37971521

ABSTRACT

Mitochondrial translation occurs on the mitochondrial ribosome, also known as the mitoribosome. The assembly of mitoribosomes is a highly coordinated process. During mitoribosome biogenesis, various assembly factors transiently associate with the nascent ribosome, facilitating the accurate and efficient construction of the mitoribosome. However, the specific factors involved in the assembly process, the precise mechanisms, and the cellular compartments involved in this vital process are not yet fully understood. In this study, we discovered a crucial role for GTP-binding protein 8 (GTPBP8) in the assembly of the mitoribosomal large subunit (mt-LSU) and mitochondrial translation. GTPBP8 is identified as a novel GTPase located in the matrix and peripherally bound to the inner mitochondrial membrane. Importantly, GTPBP8 is specifically associated with the mt-LSU during its assembly. Depletion of GTPBP8 leads to an abnormal accumulation of mt-LSU, indicating that GTPBP8 is critical for proper mt-LSU assembly. Furthermore, the absence of GTPBP8 results in reduced levels of fully assembled 55S monosomes. This impaired assembly leads to compromised mitochondrial translation and, consequently, impaired mitochondrial function. The identification of GTPBP8 as an important player in these processes provides new insights into the molecular mechanisms underlying mitochondrial protein synthesis and its regulation.


Subject(s)
Mitochondria , Mitochondrial Membranes , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Ribosomes/chemistry , Mitochondrial Ribosomes/metabolism , Protein Biosynthesis , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Mitochondrial Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
14.
J Biol Chem ; 298(8): 102214, 2022 08.
Article in English | MEDLINE | ID: mdl-35779633

ABSTRACT

Mitochondrial translation is a highly regulated process, and newly synthesized mitochondrial products must first associate with several nuclear-encoded auxiliary factors to form oxidative phosphorylation complexes. The output of mitochondrial products should therefore be in stoichiometric equilibrium with the nuclear-encoded products to prevent unnecessary energy expense or the accumulation of pro-oxidant assembly modules. In the mitochondrial DNA of Saccharomyces cerevisiae, COX1 encodes subunit 1 of the cytochrome c oxidase and COB the central core of the cytochrome bc1 electron transfer complex; however, factors regulating the expression of these mitochondrial products are not completely described. Here, we identified Mrx9p as a new factor that controls COX1 and COB expression. We isolated MRX9 in a screen for mitochondrial factors that cause poor accumulation of newly synthesized Cox1p and compromised transition to the respiratory metabolism. Northern analyses indicated lower levels of COX1 and COB mature mRNAs accompanied by an accumulation of unprocessed transcripts in the presence of excess Mrx9p. In a strain devoid of mitochondrial introns, MRX9 overexpression did not affect COX1 and COB translation or respiratory adaptation, implying Mrx9p regulates processing of COX1 and COB RNAs. In addition, we found Mrx9p was localized in the mitochondrial inner membrane, facing the matrix, as a portion of it cosedimented with mitoribosome subunits and its removal or overexpression altered Mss51p sedimentation. Finally, we showed accumulation of newly synthesized Cox1p in the absence of Mrx9p was diminished in cox14 null mutants. Taken together, these data indicate a regulatory role of Mrx9p in COX1 RNA processing.


Subject(s)
Electron Transport Complex III/genetics , Electron Transport Complex IV/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , RNA/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
15.
J Transl Med ; 21(1): 512, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37507746

ABSTRACT

Mitochondria are the only organelles regulated by two genomes. The coordinated translation of nuclear DNA (nDNA) and mitochondrial DNA (mtDNA), which together co-encode the subunits of the oxidative phosphorylation (OXPHOS) complex, is critical for determining the metabolic plasticity of tumor cells. RNA-binding protein (RBP) is a post-transcriptional regulatory factor that plays a pivotal role in determining the fate of mRNA. RBP rapidly and effectively reshapes the mitochondrial proteome in response to intracellular and extracellular stressors, mediating the cytoplasmic and mitochondrial translation balance to adjust mitochondrial respiratory capacity and provide energy for tumor cells to adapt to different environmental pressures and growth needs. This review highlights the ability of RBPs to use liquid-liquid phase separation (LLPS) as a platform for translation regulation, integrating nuclear-mitochondrial positive and retrograde signals to coordinate cross-department translation, reshape mitochondrial energy metabolism, and promote the development and survival of tumor cells.


Subject(s)
Genome, Mitochondrial , Genome, Mitochondrial/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mitochondria/genetics , Mitochondria/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Cell Nucleus/metabolism
16.
J Autoimmun ; 139: 103091, 2023 09.
Article in English | MEDLINE | ID: mdl-37595410

ABSTRACT

Obesity-induced chronic inflammation has been linked to several autoimmune diseases, including rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. The underlying mechanisms are not yet fully understood, but it is believed that chronic inflammation in adipose tissue can lead to the production of pro-inflammatory cytokines and chemokines, which can trigger immune responses and contribute to the development of autoimmune diseases. However, the underlying mechanisms that lead to the infiltration of immune cells into adipose tissue are not fully understood. In this study, we observed a time-dependent response to a high-fat diet in the liver and epididymal white adipose tissue using gene set enrichment analysis. Our findings revealed a correlation between early abnormal innate immune responses in the liver and late inflammatory response in the adipose tissue, that eventually leads to systemic inflammation. Specifically, our data suggest that the dysregulated NADH homeostasis in the mitochondrial matrix, interacting with the mitochondrial translation process, could serve as a sign marking the transition from liver inflammation to adipose tissue inflammation. Taken together, our study provides valuable insights into the molecular mechanisms underlying the development of chronic inflammation and associated autoimmune diseases in obesity.


Subject(s)
Autoimmune Diseases , Diet, High-Fat , Animals , Mice , Diet, High-Fat/adverse effects , Liver , Inflammation , Adipose Tissue , Obesity
17.
BMC Biol ; 20(1): 12, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34996455

ABSTRACT

BACKGROUND: The establishment and maintenance of functional neural connections relies on appropriate distribution and localization of mitochondria in neurites, as these organelles provide essential energy and metabolites. In particular, mitochondria are transported to axons and support local energy production to maintain energy-demanding neuronal processes including axon branching, growth, and regeneration. Additionally, local protein synthesis is required for structural and functional changes in axons, with nuclear-encoded mitochondrial mRNAs having been found localized in axons. However, it remains unclear whether these mRNAs are locally translated and whether the potential translated mitochondrial proteins are involved in the regulation of mitochondrial functions in axons. Here, we aim to further understand the purpose of such compartmentalization by focusing on the role of mitochondrial initiation factor 3 (mtIF3), whose nuclear-encoded transcripts have been shown to be present in axonal growth cones. RESULTS: We demonstrate that brain-derived neurotrophic factor (BDNF) induces local translation of mtIF3 mRNA in axonal growth cones. Subsequently, mtIF3 protein is translocated into axonal mitochondria and promotes mitochondrial translation as assessed by our newly developed bimolecular fluorescence complementation sensor for the assembly of mitochondrial ribosomes. We further show that BDNF-induced axonal growth requires mtIF3-dependent mitochondrial translation in distal axons. CONCLUSION: We describe a previously unknown function of mitochondrial initiation factor 3 (mtIF3) in axonal protein synthesis and development. These findings provide insight into the way neurons adaptively control mitochondrial physiology and axonal development via local mtIF3 translation.


Subject(s)
Axons , Brain-Derived Neurotrophic Factor , Brain-Derived Neurotrophic Factor/metabolism , Neurons/physiology , Peptide Initiation Factors/metabolism , Protein Biosynthesis
18.
J Biol Chem ; 297(1): 100869, 2021 07.
Article in English | MEDLINE | ID: mdl-34119521

ABSTRACT

Pentatricopeptide repeat (PPR) proteins are a large family of proteins that act primarily at different posttranscriptional steps of organellar gene expression. We have previously found that the Schizosaccharomyces pombe PPR protein mpal10 interacts with mitochondrial translational activator Mpa1, and both are essential for mitochondrial protein synthesis. However, it is unclear how these two proteins function in mitochondrial protein synthesis in S. pombe. In this study, we further investigated the role of Ppr10 and Mpa1 in mitochondrial protein synthesis. Mitochondrial translational initiation requires two initiation factors, Mti2 and Mti3, which bind to the small subunit of the mitochondrial ribosome (mt-SSU) during the formation of the mitochondrial translational initiation complex. Using sucrose gradient sedimentation analysis, we found that disruption of ppr10, mpa1, or the PPR motifs in Ppr10 impairs the association of Mti2 and Mti3 with the mt-SSU, suggesting that both Ppr10 and Mpa1 may be required for the interaction of Mti2 and Mti3 with the mt-SSU during the assembly of mitochondrial translational initiation complex. Loss of Ppr10 perturbs the association of mitochondrially encoded cytochrome b (cob1) and cytochrome c oxidase subunit 1 (cox1) mRNAs with assembled mitochondrial ribosomes. Proteomic analysis revealed that a fraction of Ppr10 and Mpa1 copurified with a subset of mitoribosomal proteins. The PPR motifs of Ppr10 are necessary for its interaction with Mpa1 and that disruption of these PPR motifs impairs mitochondrial protein synthesis. Our results suggest that Ppr10 and Mpa1 function together to mediate mitochondrial translational initiation.


Subject(s)
Carrier Proteins/metabolism , Mitochondria/metabolism , Peptide Chain Initiation, Translational , RNA-Binding Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Eukaryotic Initiation Factors/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/metabolism , Protein Binding , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics
19.
Mol Biol Evol ; 38(8): 3445-3458, 2021 07 29.
Article in English | MEDLINE | ID: mdl-33878189

ABSTRACT

The high mutational load of mitochondrial genomes combined with their uniparental inheritance and high polyploidy favors the maintenance of deleterious mutations within populations. How cells compose and adapt to the accumulation of disadvantageous mitochondrial alleles remains unclear. Most harmful changes are likely corrected by purifying selection, however, the intimate collaboration between mitochondria- and nuclear-encoded gene products offers theoretical potential for compensatory adaptive changes. In plants, cytoplasmic male sterilities are known examples of nucleo-mitochondrial coadaptation situations in which nuclear-encoded restorer of fertility (Rf) genes evolve to counteract the effect of mitochondria-encoded cytoplasmic male sterility (CMS) genes and restore fertility. Most cloned Rfs belong to a small monophyletic group, comprising 26 pentatricopeptide repeat genes in Arabidopsis, called Rf-like (RFL). In this analysis, we explored the functional diversity of RFL genes in Arabidopsis and found that the RFL8 gene is not related to CMS suppression but essential for plant embryo development. In vitro-rescued rfl8 plantlets are deficient in the production of the mitochondrial heme-lyase complex. A complete ensemble of molecular and genetic analyses allowed us to demonstrate that the RFL8 gene has been selected to permit the translation of the mitochondrial ccmFN2 gene encoding a heme-lyase complex subunit which derives from the split of the ccmFN gene, specifically in Brassicaceae plants. This study represents thus a clear case of nuclear compensation to a lineage-specific mitochondrial genomic rearrangement in plants and demonstrates that RFL genes can be selected in response to other mitochondrial deviancies than CMS suppression.


Subject(s)
Arabidopsis/genetics , Genome, Mitochondrial , Selection, Genetic , Arabidopsis/embryology , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cytochrome c Group/metabolism , Embryonic Development , Protein Biosynthesis , RNA Splicing
20.
J Biol Chem ; 295(7): 2057-2067, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31915244

ABSTRACT

Macrolide antibiotics, such as erythromycin and josamycin, are natural polyketide products harboring 14- to 16-membered macrocyclic lactone rings to which various sugars are attached. These antibiotics are used extensively in the clinic because of their ability to inhibit bacterial protein synthesis. More recently, some macrolides have been shown to also possess anti-inflammatory and other therapeutic activities in mammalian cells. To better understand the targets and effects of this drug class in mammalian cells, we used a genome-wide shRNA screen in K562 cancer cells to identify genes that modulate cellular sensitivity to josamycin. Among the most sensitizing hits were proteins involved in mitochondrial translation and the mitochondrial unfolded protein response, glycolysis, and the mitogen-activated protein kinase signaling cascade. Further analysis revealed that cells treated with josamycin or other antibacterial agents exhibited impaired oxidative phosphorylation and metabolic shifts to glycolysis. Interestingly, we observed that knockdown of the mitogen-activated protein kinase kinase kinase 4 (MAP3K4) gene, which contributes to p38 mitogen-activated protein kinase signaling, sensitized cells only to josamycin but not to other antibacterial agents. There is a growing interest in better characterizing the therapeutic effects and toxicities of antibiotics in mammalian cells to guide new applications in both cellular and clinical studies. To our knowledge, this is the first report of an unbiased genome-wide screen to investigate the effects of a clinically used antibiotic on human cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , MAP Kinase Kinase Kinase 4/genetics , Protein Biosynthesis/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Anti-Bacterial Agents/adverse effects , Drug Resistance, Microbial/drug effects , Erythromycin/adverse effects , Erythromycin/pharmacology , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Humans , Josamycin/adverse effects , Josamycin/pharmacology , K562 Cells , MAP Kinase Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Macrolides/adverse effects , Macrolides/pharmacology , Mitogen-Activated Protein Kinases/genetics , Oxidative Phosphorylation/drug effects , Protein Synthesis Inhibitors/adverse effects , Protein Synthesis Inhibitors/pharmacology , Unfolded Protein Response/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL