Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34287642

ABSTRACT

The ability to adapt to environmental stress, including therapeutic insult, contributes to tumor evolution and drug resistance. In suboptimal conditions, the integrated stress response (ISR) promotes survival by dampening cytosolic translation. We show that ISR-dependent survival also relies on a concomitant up-regulation of mitochondrial protein synthesis, a vulnerability that can be exploited using mitoribosome-targeting antibiotics. Accordingly, such agents sensitized to MAPK inhibition, thus preventing the development of resistance in BRAFV600E melanoma models. Additionally, this treatment compromised the growth of melanomas that exhibited elevated ISR activity and resistance to both immunotherapy and targeted therapy. In keeping with this, pharmacological inactivation of ISR, or silencing of ATF4, rescued the antitumoral response to the tetracyclines. Moreover, a melanoma patient exposed to doxycycline experienced complete and long-lasting response of a treatment-resistant lesion. Our study indicates that the repurposing of mitoribosome-targeting antibiotics offers a rational salvage strategy for targeted therapy in BRAF mutant melanoma and a therapeutic option for NRAS-driven and immunotherapy-resistant tumors.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Mitochondrial Ribosomes/drug effects , Aged , Animals , Cell Line, Tumor , Doxycycline/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Humans , Male , Melanoma/genetics , Melanoma/mortality , Mice, Inbred C57BL , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Stress, Physiological/drug effects , Tigecycline/pharmacology , Uveal Neoplasms/drug therapy , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Nat Commun ; 12(1): 3607, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34127662

ABSTRACT

Ribosomes are recycled for a new round of translation initiation by dissociation of ribosomal subunits, messenger RNA and transfer RNA from their translational post-termination complex. Here we present cryo-EM structures of the human 55S mitochondrial ribosome (mitoribosome) and the mitoribosomal large 39S subunit in complex with mitoribosome recycling factor (RRFmt) and a recycling-specific homolog of elongation factor G (EF-G2mt). These structures clarify an unusual role of a mitochondria-specific segment of RRFmt, identify the structural distinctions that confer functional specificity to EF-G2mt, and show that the deacylated tRNA remains with the dissociated 39S subunit, suggesting a distinct sequence of events in mitoribosome recycling. Furthermore, biochemical and structural analyses reveal that the molecular mechanism of antibiotic fusidic acid resistance for EF-G2mt is markedly different from that of mitochondrial elongation factor EF-G1mt, suggesting that the two human EF-Gmts have evolved diversely to negate the effect of a bacterial antibiotic.


Subject(s)
Drug Resistance, Microbial/genetics , Mitochondrial Ribosomes/chemistry , Mitochondrial Ribosomes/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Cryoelectron Microscopy , Humans , Mitochondria , Mitochondrial Ribosomes/drug effects , Models, Molecular , Peptide Elongation Factor G/chemistry , Peptide Elongation Factor G/genetics , Peptide Elongation Factor G/metabolism , Protein Conformation , Protein Conformation, alpha-Helical , Protein Domains , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics
3.
Commun Biol ; 4(1): 300, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33686140

ABSTRACT

Mitochondrial translation appears to involve two stalled-ribosome rescue factors (srRFs). One srRF is an ICT1 protein from humans that rescues a "non-stop" type of mitochondrial ribosomes (mitoribosomes) stalled on mRNA lacking a stop codon, while the other, C12orf65, reportedly has functions that overlap with those of ICT1; however, its primary role remains unclear. We herein demonstrated that the Saccharomyces cerevisiae homolog of C12orf65, Pth3 (Rso55), preferentially rescued antibiotic-dependent stalled mitoribosomes, which appear to represent a "no-go" type of ribosomes stalled on intact mRNA. On media containing a non-fermentable carbon source, which requires mitochondrial gene expression, respiratory growth was impaired significantly more by the deletion of PTH3 than that of the ICT1 homolog PTH4 in the presence of antibiotics that inhibit mitochondrial translation, such as tetracyclines and macrolides. Additionally, the in organello labeling of mitochondrial translation products and quantification of mRNA levels by quantitative RT-PCR suggested that in the presence of tetracycline, the deletion of PTH3, but not PTH4, reduced the protein expression of all eight mtDNA-encoded genes at the post-transcriptional or translational level. These results indicate that Pth3 can function as a mitochondrial srRF specific for ribosomes stalled by antibiotics and plays a role in antibiotic resistance in fungi.


Subject(s)
Anti-Bacterial Agents/pharmacology , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/drug effects , Peptide Termination Factors/metabolism , Protein Biosynthesis/drug effects , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Drug Resistance, Fungal , Gene Expression Regulation, Fungal , Mitochondrial Proteins/genetics , Mitochondrial Ribosomes/metabolism , Mutation , Peptide Termination Factors/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
4.
Nucleic Acids Res ; 49(1): 371-382, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33300043

ABSTRACT

Mammalian mitochondria have their own dedicated protein synthesis system, which produces 13 essential subunits of the oxidative phosphorylation complexes. We have reconstituted an in vitro translation system from mammalian mitochondria, utilizing purified recombinant mitochondrial translation factors, 55S ribosomes from pig liver mitochondria, and a tRNA mixture from either Escherichia coli or yeast. The system is capable of translating leaderless mRNAs encoding model proteins (DHFR and nanoLuciferase) or some mtDNA-encoded proteins. We show that a leaderless mRNA, encoding nanoLuciferase, is faithfully initiated without the need for any auxiliary factors other than IF-2mt and IF-3mt. We found that the ribosome-dependent GTPase activities of both the translocase EF-G1mt and the recycling factor EF-G2mt are insensitive to fusidic acid (FA), the translation inhibitor that targets bacterial EF-G homologs, and consequently the system is resistant to FA. Moreover, we demonstrate that a polyproline sequence in the protein causes 55S mitochondrial ribosome stalling, yielding ribosome nascent chain complexes. Analyses of the effects of the Mg concentration on the polyproline-mediated ribosome stalling suggested the unique regulation of peptide elongation by the mitoribosome. This system will be useful for analyzing the mechanism of translation initiation, and the interactions between the nascent peptide chain and the mitochondrial ribosome.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Ribosomes/metabolism , Peptides/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , 5' Untranslated Regions , Animals , Cell-Free System , DNA/chemical synthesis , Escherichia coli , Eukaryotic Initiation Factors/metabolism , Humans , Luciferases/biosynthesis , Luciferases/genetics , Magnesium/pharmacology , Mitochondrial Proteins/genetics , Mitochondrial Ribosomes/drug effects , Mitochondrial Ribosomes/ultrastructure , Oxidative Phosphorylation , Peptide Chain Initiation, Translational , Peptide Elongation Factors/physiology , Peptides/genetics , Protein Biosynthesis/drug effects , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Swine , Tetrahydrofolate Dehydrogenase/biosynthesis , Tetrahydrofolate Dehydrogenase/genetics
5.
Proc Natl Acad Sci U S A ; 117(47): 29851-29861, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168716

ABSTRACT

Kinetoplastids are unicellular eukaryotic parasites responsible for such human pathologies as Chagas disease, sleeping sickness, and leishmaniasis. They have a single large mitochondrion, essential for the parasite survival. In kinetoplastid mitochondria, most of the molecular machineries and gene expression processes have significantly diverged and specialized, with an extreme example being their mitochondrial ribosomes. These large complexes are in charge of translating the few essential mRNAs encoded by mitochondrial genomes. Structural studies performed in Trypanosoma brucei already highlighted the numerous peculiarities of these mitoribosomes and the maturation of their small subunit. However, several important aspects mainly related to the large subunit (LSU) remain elusive, such as the structure and maturation of its ribosomal RNA. Here we present a cryo-electron microscopy study of the protozoans Leishmania tarentolae and Trypanosoma cruzi mitoribosomes. For both species, we obtained the structure of their mature mitoribosomes, complete rRNA of the LSU, as well as previously unidentified ribosomal proteins. In addition, we introduce the structure of an LSU assembly intermediate in the presence of 16 identified maturation factors. These maturation factors act on both the intersubunit and the solvent sides of the LSU, where they refold and chemically modify the rRNA and prevent early translation before full maturation of the LSU.


Subject(s)
Leishmania/physiology , Mitochondrial Ribosomes/ultrastructure , RNA Processing, Post-Transcriptional/physiology , Ribosome Subunits, Large, Eukaryotic/metabolism , Trypanosoma cruzi/physiology , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Chagas Disease/drug therapy , Chagas Disease/parasitology , Cryoelectron Microscopy , Humans , Leishmania/cytology , Leishmania/drug effects , Leishmaniasis/drug therapy , Leishmaniasis/parasitology , Mitochondrial Ribosomes/drug effects , Mitochondrial Ribosomes/metabolism , Models, Molecular , RNA Processing, Post-Transcriptional/drug effects , RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/ultrastructure , Trypanosoma cruzi/cytology , Trypanosoma cruzi/drug effects
6.
Aging (Albany NY) ; 12(11): 10162-10179, 2020 05 24.
Article in English | MEDLINE | ID: mdl-32452826

ABSTRACT

Cancer stem cells (CSCs) have been proposed to be responsible for tumor recurrence, distant metastasis and drug-resistance, in the vast majority of cancer patients. Therefore, there is an urgent need to identify new drugs that can target and eradicate CSCs. To identify new molecular targets that are unique to CSCs, we previously compared MCF7 2D-monolayers with 3D-mammospheres, which are enriched in CSCs. We observed that 25 mitochondrial-related proteins were >100-fold over-expressed in 3D-mammospheres. Here, we used these 25 proteins to derive short gene signatures to predict distant metastasis (in N=1,395 patients) and tumor recurrence (in N=3,082 patients), by employing a large collection of transcriptional profiling data from ER(+) breast cancer patients. This analysis resulted in a 4-gene signature for predicting distant metastasis, with a hazard ratio of 1.91-fold (P=2.2e-08). This provides clinical evidence to support a role for CSC mitochondria in metastatic dissemination. Next, we employed a panel of mitochondrial inhibitors, previously shown to target mitochondria and selectively inhibit 3D-mammosphere formation in MCF7 cells and cell migration in MDA-MB-231 cells. Remarkably, these five mitochondrial inhibitors had only minor effects or no effect on MDA-MB-231 tumor formation, but preferentially and selectively inhibited tumor cell metastasis, without causing significant toxicity. Mechanistically, all five mitochondrial inhibitors have been previously shown to induce ATP-depletion in cancer cells. Since 3 of these 5 inhibitors were designed to target the large mitochondrial ribosome, we next interrogated whether genes encoding the large mitochondrial ribosomal proteins (MRPL) also show prognostic value in the prediction of distant metastasis in both ER(+) and ER(-) breast cancer patients. Interestingly, gene signatures composed of 6 to 9 MRPL mRNA-transcripts were indeed sufficient to predict distant metastasis, tumor recurrence and Tamoxifen resistance. These gene signatures could be useful as companion diagnostics to assess which patients may benefit most from anti-mito-ribosome therapy. Overall, our studies provide the necessary proof-of-concept, and in vivo functional evidence, that mitochondrial inhibitors can successfully and selectively target the biological process of cancer cell metastasis. Ultimately, we envision that mitochondrial inhibitors could be employed to develop new treatment protocols, for clinically providing metastasis prophylaxis, to help prevent poor clinical outcomes in cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Mitochondria/drug effects , Mitochondrial Ribosomes/drug effects , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Humans , MCF-7 Cells , Mitochondria/metabolism , Mitochondrial Proteins/analysis , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/metabolism , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/genetics , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Prognosis , Proof of Concept Study , Ribosomal Proteins/analysis , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Spheroids, Cellular
7.
Expert Opin Drug Saf ; 16(7): 833-843, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28538105

ABSTRACT

INTRODUCTION: Linezolid inhibits bacterial growth by targeting bacterial ribosomes and by interfering with bacterial protein synthesis. Lactic acidosis is a rare, but potentially lethal, side effect of linezolid. Areas covered: The pathogenesis of linezolid-induced lactic acidosis is reviewed with special emphasis on aspects relevant to the recognition, prevention and treatment of the syndrome. Expert opinion: Linezolid-induced lactic acidosis reflects the untoward interaction between the drug and mitochondrial ribosomes. The inhibition of mitochondrial protein synthesis diminishes the respiratory chain enzyme content and thus limits aerobic energy production. As a result, anaerobic glycolysis and lactate generation accelerate independently from tissue hypoxia. In the absence of any confirmatory test, linezolid-induced lactic acidosis should be suspected only after exclusion of other, more common, causes of lactic acidosis such as hypoxemia, anemia or low cardiac output. Normal-to-high whole-body oxygen delivery, high venous oxygen saturation and lack of response to interventions that effectively increase tissue oxygen provision all suggest a primary defect in oxygen use at the mitochondrial level. During prolonged therapy with linezolid, blood drug and lactate levels should be regularly monitored. The current standard-of-care treatment of linezolid-induced lactic acidosis consists of drug withdrawal to reverse mitochondrial intoxication and intercurrent life support.


Subject(s)
Acidosis, Lactic/chemically induced , Anti-Bacterial Agents/administration & dosage , Linezolid/administration & dosage , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Glycolysis/drug effects , Humans , Linezolid/adverse effects , Linezolid/pharmacology , Mitochondrial Ribosomes/drug effects , Oxygen/metabolism , Ribosomes/metabolism
8.
Nat Rev Clin Oncol ; 14(1): 11-31, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27141887

ABSTRACT

Awareness that the metabolic phenotype of cells within tumours is heterogeneous - and distinct from that of their normal counterparts - is growing. In general, tumour cells metabolize glucose, lactate, pyruvate, hydroxybutyrate, acetate, glutamine, and fatty acids at much higher rates than their nontumour equivalents; however, the metabolic ecology of tumours is complex because they contain multiple metabolic compartments, which are linked by the transfer of these catabolites. This metabolic variability and flexibility enables tumour cells to generate ATP as an energy source, while maintaining the reduction-oxidation (redox) balance and committing resources to biosynthesis - processes that are essential for cell survival, growth, and proliferation. Importantly, experimental evidence indicates that metabolic coupling between cell populations with different, complementary metabolic profiles can induce cancer progression. Thus, targeting the metabolic differences between tumour and normal cells holds promise as a novel anticancer strategy. In this Review, we discuss how cancer cells reprogramme their metabolism and that of other cells within the tumour microenvironment in order to survive and propagate, thus driving disease progression; in particular, we highlight potential metabolic vulnerabilities that might be targeted therapeutically.


Subject(s)
Neoplasms/metabolism , Acetyl Coenzyme A/metabolism , Adaptation, Physiological , Amino Acids/metabolism , Antineoplastic Agents/therapeutic use , Antioxidants/metabolism , Autophagy/physiology , Blood Glucose/metabolism , Energy Metabolism/drug effects , Epigenomics , Fatty Acids/metabolism , Genetic Heterogeneity , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Ketone Bodies/metabolism , Lactic Acid/metabolism , Lipids/biosynthesis , Mitochondria/drug effects , Mitochondrial Ribosomes/drug effects , Neoplasms/drug therapy , Nucleic Acids/biosynthesis , Oxidative Stress/drug effects , Pyruvic Acid/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/metabolism , Tumor Microenvironment
9.
Cell Tissue Res ; 368(1): 187-200, 2017 04.
Article in English | MEDLINE | ID: mdl-27807702

ABSTRACT

Capacitation comprises a series of structural and functional modifications of sperm that confer fertilizing ability. We previously reported that the testis-specific isoform of Na/K-ATPase (ATP1A4) regulated bovine sperm capacitation through signaling mechanisms involving kinases. During subsequent investigations to elucidate mechanisms by which ATP1A4 regulates sperm capacitation, we observed that ATP1A4 was localised in both raft and non-raft fractions of the sperm plasma membrane and that its total content was increased in both membrane fractions during capacitation. The objective of the present study was to investigate mechanism(s) of capacitation-associated increase in the content of ATP1A4. Despite the widely accepted dogma of transcriptional/translational quiescence, incubation of sperm with either ouabain (specific ligand for ATP1A4) or heparin increased ATP1A4 content in raft and non-raft sperm membrane fractions, total sperm protein extracts (immunoblotting) and fixed sperm (flow cytometry), with a concurrent increase in Na/K-ATPase enzyme activity. This capacitation-associated increase in ATP1A4 content was partially decreased by chloramphenicol (mitochondrial translation inhibitor) but not affected by actinomycin D (transcription inhibitor). To demonstrate de novo ATP1A4 synthesis, we evaluated incorporation of bodipy conjugated lysine in this protein during capacitation. A partial decrease in bodipy-lysine incorporation occurred in ATP1A4 from sperm capacitated in the presence of chloramphenicol. Therefore, increased ATP1A4 content during capacitation was attributed to mitochondrial translation of ATP1A4 mRNA present in ejaculated sperm, rather than gene transcription. To our knowledge, this is the first report demonstrating ATP1A4 synthesis during bovine sperm capacitation.


Subject(s)
Mitochondrial Ribosomes/metabolism , Protein Biosynthesis , Sodium-Potassium-Exchanging ATPase/metabolism , Sperm Capacitation , Testis/metabolism , Amino Acids/metabolism , Animals , Cattle , Chloramphenicol/pharmacology , Dactinomycin/pharmacology , Detergents/pharmacology , Flow Cytometry , Fluorescence , G(M1) Ganglioside/metabolism , Isoenzymes/metabolism , Male , Membrane Microdomains/metabolism , Mitochondrial Ribosomes/drug effects , Organ Specificity/drug effects , Phosphoproteins/metabolism , Protein Biosynthesis/drug effects , Reproducibility of Results , Solubility , Sperm Capacitation/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Testis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...