Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Nature ; 631(8020): 409-414, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961288

ABSTRACT

Bedaquiline (BDQ), a first-in-class diarylquinoline anti-tuberculosis drug, and its analogue, TBAJ-587, prevent the growth and proliferation of Mycobacterium tuberculosis by inhibiting ATP synthase1,2. However, BDQ also inhibits human ATP synthase3. At present, how these compounds interact with either M. tuberculosis ATP synthase or human ATP synthase is unclear. Here we present cryogenic electron microscopy structures of M. tuberculosis ATP synthase with and without BDQ and TBAJ-587 bound, and human ATP synthase bound to BDQ. The two inhibitors interact with subunit a and the c-ring at the leading site, c-only sites and lagging site in M. tuberculosis ATP synthase, showing that BDQ and TBAJ-587 have similar modes of action. The quinolinyl and dimethylamino units of the compounds make extensive contacts with the protein. The structure of human ATP synthase in complex with BDQ reveals that the BDQ-binding site is similar to that observed for the leading site in M. tuberculosis ATP synthase, and that the quinolinyl unit also interacts extensively with the human enzyme. This study will improve researchers' understanding of the similarities and differences between human ATP synthase and M. tuberculosis ATP synthase in terms of the mode of BDQ binding, and will allow the rational design of novel diarylquinolines as anti-tuberculosis drugs.


Subject(s)
Antitubercular Agents , Cryoelectron Microscopy , Diarylquinolines , Models, Molecular , Mycobacterium tuberculosis , Diarylquinolines/pharmacology , Diarylquinolines/chemistry , Humans , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/drug effects , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Binding Sites , Quinolines/chemistry , Quinolines/pharmacology , Protein Subunits/metabolism , Protein Subunits/chemistry , Protein Subunits/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/chemistry , Imidazoles , Piperidines , Pyridines
2.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731874

ABSTRACT

The mitochondrial protein IF1 is upregulated in many tumors and acts as a pro-oncogenic protein through its interaction with the ATP synthase and the inhibition of apoptosis. We have recently characterized the molecular nature of the IF1-Oligomycin Sensitivity Conferring Protein (OSCP) subunit interaction; however, it remains to be determined whether this interaction could be targeted for novel anti-cancer therapeutic intervention. We generated mitochondria-targeting peptides to displace IF1 from the OSCP interaction. The use of one selective peptide led to displacement of the inhibitor IF1 from ATP synthase, as shown by immunoprecipitation. NMR spectroscopy analysis, aimed at clarifying whether these peptides were able to directly bind to the OSCP protein, identified a second peptide which showed affinity for the N-terminal region of this subunit overlapping the IF1 binding region. In situ treatment with the membrane-permeable derivatives of these peptides in HeLa cells, that are silenced for the IF1 inhibitor protein, showed significant inhibition in mitochondrial permeability transition and no effects on mitochondrial respiration. These peptides mimic the effects of the IF1 inhibitor protein in cancer HeLa cells and confirm that the IF1-OSCP interaction inhibits apoptosis. A third peptide was identified which counteracts the anti-apoptotic role of IF1, showing that OSCP is a promising target for anti-cancer therapies.


Subject(s)
Mitochondria , Mitochondrial Proton-Translocating ATPases , Peptides , Humans , Apoptosis/drug effects , ATPase Inhibitory Protein/drug effects , ATPase Inhibitory Protein/metabolism , HeLa Cells , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Peptides/pharmacology , Peptides/chemistry , Peptides/metabolism , Protein Binding
3.
Mol Cell ; 57(1): 165-78, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25544562

ABSTRACT

Although endogenous siRNAs (endo-siRNAs) have been described in many species, still little is known about their endogenous utility. Here, we show that Drosophila hairpin RNAs (hpRNAs) generate an endo-siRNA class with predominant expression in testes. Although hpRNAs are universally recently evolved, we identify highly complementary protein-coding targets for all hpRNAs. Importantly, we find broad evidence for evolutionary divergences that preferentially maintain compensatory pairing between hpRNAs and targets, serving as first evidence for adaptive selection for siRNA-mediated target regulation in metazoans. We demonstrate organismal impact of hpRNA activity, since knockout of hpRNA1 derepresses its target ATP synthase-ß in testes and compromises spermatogenesis and male fertility. Moreover, we reveal surprising male-specific impact of RNAi factors on germ cell development and fertility, consistent with testis-directed function of the hpRNA pathway. Finally, the collected hpRNA loci chronicle an evolutionary timeline that reflects their origins from prospective target genes, mirroring a strategy described for plant miRNAs.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Insect Proteins/genetics , RNA, Small Interfering/genetics , Spermatogenesis/genetics , Testis/metabolism , Adaptation, Physiological/genetics , Animals , Base Sequence , Biological Evolution , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Fertility/genetics , Humans , Insect Proteins/antagonists & inhibitors , Insect Proteins/metabolism , Male , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/growth & development
4.
Pharmacol Res ; 165: 105458, 2021 03.
Article in English | MEDLINE | ID: mdl-33515708

ABSTRACT

Bone resorption by osteoclasts is an energy consuming activity, which depends on mitochondrial ATP. ATP5B, a mitochondrial ATP synthase beta subunit, is a catalytic core involved in producing ATP. Here, we investigated the contribution of ATP5B in osteoclast differentiation and joint destruction. ATP5B (LV-ATP5B) targeting or non-targeting (LV-NC) siRNA containing lentivirus particles were transduced into bone marrow macrophage derived osteoclasts or locally administered to arthritic mouse joints. Inhibition of ATP5B reduced the expression of osteoclast related genes and proteins, suppressed bone resorption by significantly impairing F-actin formation and decreased the levels of adhesion-associated proteins. In addition, ATP5B deficiency caused osteoclast mitochondrial dysfunction and, impaired the secretion of vacuole protons and MMP9. Importantly, inhibition of ATP5B expression, protected arthritis mice from joint destructions although serum levels of inflammatory mediators (TNF-α, IL-1ß) and IgG2α antibodies were unaffected. These results demonstrate an essential function of ATP5B in osteoclast differentiation and bone resorption, and suggest it as a potential therapeutic target for protecting bones in RA.


Subject(s)
Arthritis, Experimental/genetics , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Osteoclasts/physiology , Osteogenesis/genetics , RNA, Small Interfering/genetics , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/therapy , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/prevention & control , Gene Targeting/methods , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mitochondrial Proton-Translocating ATPases/biosynthesis , RNA, Small Interfering/administration & dosage
5.
Int J Mol Sci ; 22(18)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34575875

ABSTRACT

Mitochondrial dysfunction during ischemic stroke ultimately manifests as ATP depletion. Mitochondrial ATP synthase upon loss of mitochondrial membrane potential during ischemia rapidly hydrolyses ATP and thus contributes to ATP depletion. Increasing evidence suggests that inhibition of ATP synthase limits ATP depletion and is protective against ischemic tissue damage. Bedaquiline (BDQ) is an anti-microbial agent, approved for clinical use, that inhibits ATP synthase of Mycobacteria; however recently it has been shown to act on mitochondrial ATP synthase, inhibiting both ATP synthesis and hydrolysis in low micromolar concentrations. In this study, we investigated whether preconditioning with BDQ can alleviate ischemia/reperfusion-induced brain injury in Wistar rats after middle cerebral artery occlusion-reperfusion and whether it affects mitochondrial functions. We found that BDQ was effective in limiting necrosis and neurological dysfunction during ischemia-reperfusion. BDQ also caused inhibition of ATPase activity, mild uncoupling of respiration, and stimulated mitochondrial respiration both in healthy and ischemic mitochondria. Mitochondrial calcium retention capacity was unaffected by BDQ preconditioning. We concluded that BDQ has neuroprotective properties associated with its action on mitochondrial respiration and ATPase activity.


Subject(s)
Diarylquinolines/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Reperfusion Injury/metabolism , Stroke/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Respiration/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotection/drug effects , Rats , Reperfusion Injury/drug therapy , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Stroke/drug therapy , Stroke/etiology , Stroke/pathology
6.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360894

ABSTRACT

Disruption of retinal pigment epithelial (RPE) barrier integrity is involved in the pathology of several blinding retinal diseases including age-related macular degeneration (AMD) and diabetic retinopathy (DR), but the underlying causes and pathophysiology are not completely well-defined. Mitochondria dysfunction has often been considered as a potential candidate implicated in such a process. In this study, we aimed to dissect the role of different mitochondrial components; specifically, those of oxidative phosphorylation (OxPhos), in maintaining the barrier functionality of RPE. Electric cell-substrate impedance sensing (ECIS) technology was used to collect multi-frequency electrical impedance data to assess in real-time the barrier formation of the RPE cells. For this purpose, the human retinal pigment epithelial cell line-ARPE-19-was used and treated with varying concentrations of specific mitochondrial inhibitors that target different steps in OxPhos: Rotenone for complex I (the largest protein complex in the electron transport chain (ETC)); oligomycin for ATP synthase; and carbonyl cyanide-p-trifluoromethoxyphenyl hydrazone (FCCP) for uncoupling ATP synthesis from the accompanying ETC. Furthermore, data were modeled using the ECIS-Zθ software to investigate in depth the effects of these inhibitors on three separate barrier parameters: cell-cell interactions (Rb), cell-matrix interactions (α), and the cell membrane capacitance (Cm). The viability of ARPE-19 cells was determined by lactate dehydrogenase (LDH) Cytotoxicity Assay. The ECIS program's modeling demonstrated that FCCP and thus OxPhos uncoupling disrupt the barrier function in the ARPE-19 cells across all three components of the total resistance (Rb, α, and Cm) in a dose-dependent manner. On the other hand, oligomycin and thus ATP synthase inhibition mostly affects the ARPE-19 cells' attachment to their substrate evident by a significant decrease in α resistance in a dose-dependent manner, both at the end and throughout the duration of the experiment. On the contrary, rotenone and complex I inhibition mostly affect the ARPE-19 paracellular resistance Rb in a dose-dependent manner compared to basolateral resistance α or Cm. Our results clearly demonstrate differential roles for different mitochondrial components in maintaining RPE cell functionality in which uncoupling of OxPhos is a major contributing factor to the disruption barrier function. Such differences can be used in investigating gene expression as well as for screening of selective agents that improve the OxPhos coupling efficiency to be used in the therapeutic approach for treating RPE-related retinal diseases.


Subject(s)
Blood-Retinal Barrier/metabolism , Diabetic Retinopathy/metabolism , Epithelial Cells/metabolism , Macular Degeneration/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Retinal Pigment Epithelium/metabolism , Blood-Retinal Barrier/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacokinetics , Cell Line , Cell Survival/drug effects , Electric Impedance , Electron Transport/drug effects , Enzyme Inhibitors/pharmacokinetics , Humans , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Oligomycins/pharmacokinetics , Retinal Pigment Epithelium/drug effects , Rotenone/pharmacokinetics
7.
Int J Cancer ; 146(12): 3474-3484, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32144767

ABSTRACT

Modulation of prostate stromal cells (PrSCs) within tumor tissues is gaining attention for the treatment of solid tumors. Using our original in vitro coculture system, we previously reported that leucinostatin (LCS)-A, a peptide mycotoxin, inhibited prostate cancer DU-145 cell growth through reduction of insulin-like growth factor 1 (IGF-I) expression in PrSCs. To further obtain additional bioactive compounds from LCS-A, we designed and synthesized a series of LCS-A derivatives as compounds that target PrSCs. Among the synthesized LCS-A derivatives, LCS-7 reduced IGF-I expression in PrSCs with lower toxicity to PrSCs and mice than LCS-A. As LCS-A has been suggested to interact with mitochondrial adenosine triphosphate (ATP) synthase, a docking study was performed to elucidate the mechanism of reduced IGF-I expression in the PrSCs. As expected, LCS-A and LCS-7 directly interacted with mitochondrial ATP synthase, and like LCS-A and LCS-7, other mitochondrial ATP synthase inhibitors also reduced the expression of IGF-I by PrSCs. Furthermore, LCS-A and LCS-7 significantly decreased the growth of mouse xenograft tumors. Based on these data, we propose that the mitochondrial ATP synthases-IGF-I axis of PrSCs plays a critical role on cancer cell growth and inhibition could be a potential anticancer target for prostate cancer.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Insulin-Like Growth Factor I/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Stromal Cells/drug effects , Animals , Antimicrobial Cationic Peptides/therapeutic use , Cell Line, Tumor , Coculture Techniques , Female , Humans , Male , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Molecular Docking Simulation , Prostate/cytology , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
8.
Breast Cancer Res Treat ; 176(2): 271-289, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31006104

ABSTRACT

PURPOSE: Primary breast and prostate cancers can be cured, but metastatic disease cannot. Identifying cell factors that predict metastatic potential could guide both prognosis and treatment. METHODS: We used Cell-SELEX to screen an RNA aptamer library for differential binding to prostate cancer cell lines with high vs. low metastatic potential. Mass spectroscopy, immunoblot, and immunohistochemistry were used to identify and validate aptamer targets. Aptamer properties were tested in vitro, in xenograft models, and in clinical biopsies. Gene expression datasets were queried for target associations in cancer. RESULTS: We identified a novel aptamer (Apt63) that binds to the beta subunit of F1Fo ATP synthase (ATP5B), present on the plasma membrane of certain normal and cancer cells. Apt63 bound to plasma membranes of multiple aggressive breast and prostate cell lines, but not to normal breast and prostate epithelial cells, and weakly or not at all to non-metastasizing cancer cells; binding led to rapid cell death. A single intravenous injection of Apt63 induced rapid, tumor cell-selective binding and cytotoxicity in MDA-MB-231 xenograft tumors, associated with endonuclease G nuclear translocation and DNA fragmentation. Apt63 was not toxic to non-transformed epithelial cells in vitro or adjacent normal tissue in vivo. In breast cancer tissue arrays, plasma membrane staining with Apt63 correlated with tumor stage (p < 0.0001, n = 416) and was independent of other cancer markers. Across multiple datasets, ATP5B expression was significantly increased relative to normal tissue, and negatively correlated with metastasis-free (p = 0.0063, 0.00039, respectively) and overall (p = 0.050, 0.0198) survival. CONCLUSION: Ecto-ATP5B binding by Apt63 may disrupt an essential survival mechanism in a subset of tumors with high metastatic potential, and defines a novel category of cancers with potential vulnerability to ATP5B-targeted therapy. Apt63 is a unique tool for elucidating the function of surface ATP synthase, and potentially for predicting and treating metastatic breast and prostate cancer.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Breast Neoplasms/pathology , Cell Membrane/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Prostatic Neoplasms/pathology , Administration, Intravenous , Animals , Aptamers, Nucleotide/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Early Detection of Cancer , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Male , Mice , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Neoplasm Staging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , SELEX Aptamer Technique , Treatment Outcome , Up-Regulation , Xenograft Model Antitumor Assays
9.
Physiol Plant ; 167(2): 188-204, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30467859

ABSTRACT

Research has begun to elucidate the signal transduction pathway(s) that control cellular responses to changes in mitochondrial status. Important tools in such studies are chemical inhibitors used to initiate mitochondrial dysfunction. This study compares the effect of different inhibitors and treatment conditions on the transcript amount of nuclear genes specifically responsive to mitochondrial dysfunction in leaf of Nicotiana tabacum L. cv. Petit Havana. The Complex III inhibitors antimycin A (AA) and myxothiazol (MYXO), and the Complex V inhibitor oligomycin (OLIGO), each increased the transcript amount of the mitochondrial dysfunction genes. Transcript responses to OLIGO were greater during treatment in the dark than in the light, and the dark treatment resulted in cell death. In the dark, transcript responses to AA and MYXO were similar to one another, despite MYXO leading to cell death. In the light, transcript responses to AA and MYXO diverged, despite cell viability remaining high with either inhibitor. This divergent response may be due to differential signaling from the chloroplast because only AA also inhibited cyclic electron transport, resulting in a strong acceptor-side limitation in photosystem I. In the light, chemical inhibition of chloroplast electron transport reduced transcript responses to AA, while having no effect on the response to MYXO, and increasing the response to OLIGO. Hence, when studying mitochondrial dysfunction signaling, different inhibitor and treatment combinations differentially affect linked processes (e.g. chloroplast function and cell fate) that then contribute to measured responses. Therefore, inhibitor and treatment conditions should be chosen to align with specific study goals.


Subject(s)
Chloroplasts/metabolism , Mitochondria/metabolism , Nicotiana/genetics , Signal Transduction , Antimycin A/pharmacology , Chloroplasts/radiation effects , Electron Transport/drug effects , Electron Transport Complex III/antagonists & inhibitors , Light , Methacrylates/pharmacology , Mitochondria/radiation effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Oligomycins/pharmacology , Photosystem I Protein Complex/drug effects , Photosystem I Protein Complex/genetics , Photosystem I Protein Complex/metabolism , Plant Leaves/genetics , Plant Leaves/physiology , Plant Leaves/radiation effects , Thiazoles/pharmacology , Nicotiana/physiology , Nicotiana/radiation effects
10.
Anal Bioanal Chem ; 411(26): 6899-6911, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31407049

ABSTRACT

Adenosine triphosphate (ATP) is the main energy source in cells and an important biomolecule participating in cellular reactions in living organisms. Since the ATP level changes dynamically reflecting the development of a debilitating disease or carcinogenesis, we have focused in this work on monitoring of the oligomycin (OMC)-modulated ATP synthase inhibition using a fluorescent-switching DNA aptamer designed for the detection of ATP (Apt(ATP)), as the model for studies of dynamic ATP level variation. The behavior of the ATP aptamer has been characterized using fluorescence spectroscopy. The Intramolecular fluorescence resonance energy transfer (iFRET) operates in the proposed aptamer from the FAM dye moiety to guanines of the aptamer G-quadruplex when the target ATP is present and binds to the aptamer changing its conformation. The iFRET process enables the detection of ATP down to the limit of detection, LOD = 17 µM, without resorting to any extra chemi-amplification schemes. The selectivity coefficients for relevant interferent triphosphates (UTP, GTP, and CTP) are low for the same concentration as that of ATP. We have demonstrated an efficient transfection of intact cells and OMC-treated SW480 colon cancer cells with Apt(ATP), using microscopic imaging, iFRET measurements, and cell viability testing with MTT method. The applicability of the switching DNA aptamer for the analysis of real samples, obtained by lysis of SW480 cells, was also tested. The proposed Apt(ATP) may be considered as a viable candidate for utilization in measurements of dynamic ATP level modulation in cells in different stages of cancer development and testing of new drugs in pharmacological studies. Graphical abstract.


Subject(s)
Adenosine Triphosphate/analysis , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer/methods , Adenosine Triphosphate/metabolism , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , G-Quadruplexes , Humans , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasms/metabolism , Oligomycins/pharmacology
11.
PLoS Genet ; 12(7): e1006161, 2016 07.
Article in English | MEDLINE | ID: mdl-27442014

ABSTRACT

Most of the ATP in living cells is produced by an F-type ATP synthase. This enzyme uses the energy of a transmembrane electrochemical proton gradient to synthesize ATP from ADP and inorganic phosphate. Proton movements across the membrane domain (FO) of the ATP synthase drive the rotation of a ring of 8-15 c-subunits, which induces conformational changes in the catalytic part (F1) of the enzyme that ultimately promote ATP synthesis. Two paralogous nuclear genes, called Atp9-5 and Atp9-7, encode structurally different c-subunits in the filamentous fungus Podospora anserina. We have in this study identified differences in the expression pattern for the two genes that correlate with the mitotic activity of cells in vegetative mycelia: Atp9-7 is transcriptionally active in non-proliferating (stationary) cells while Atp9-5 is expressed in the cells at the extremity (apex) of filaments that divide and are responsible for mycelium growth. When active, the Atp9-5 gene sustains a much higher rate of c-subunit synthesis than Atp9-7. We further show that the ATP9-7 and ATP9-5 proteins have antagonist effects on the longevity of P. anserina. Finally, we provide evidence that the ATP9-5 protein sustains a higher rate of mitochondrial ATP synthesis and yield in ATP molecules per electron transferred to oxygen than the c-subunit encoded by Atp9-7. These findings reveal that the c-subunit genes play a key role in the modulation of ATP synthase production and activity along the life cycle of P. anserina. Such a degree of sophistication for regulating aerobic energy metabolism has not been described before.


Subject(s)
Energy Metabolism , Fungal Proteins/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Podospora/genetics , Aerobiosis , Enzyme Inhibitors/pharmacology , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/metabolism , Gene Expression , Gene Expression Regulation, Fungal , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/metabolism , Oligomycins/pharmacology , Podospora/enzymology , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism
12.
Biochim Biophys Acta Bioenerg ; 1859(9): 762-774, 2018 09.
Article in English | MEDLINE | ID: mdl-29886048

ABSTRACT

The ATP synthase is a reversible nanomotor that gyrates its central rotor clockwise (CW) to synthesize ATP and in counter clockwise (CCW) direction to hydrolyse it. In bacteria and mitochondria, two natural inhibitor proteins, namely the ε and IF1 subunits, prevent the wasteful CCW F1FO-ATPase activity by blocking γ rotation at the αDP/ßDP/γ interface of the F1 portion. In Paracoccus denitrificans and related α-proteobacteria, we discovered a different natural F1-ATPase inhibitor named ζ. Here we revise the functional and structural data showing that this novel ζ subunit, although being different to ε and IF1, it also binds to the αDP/ßDP/γ interface of the F1 of P. denitrificans. ζ shifts its N-terminal inhibitory domain from an intrinsically disordered protein region (IDPr) to an α-helix when inserted in the αDP/ßDP/γ interface. We showed for the first time the key role of a natural ATP synthase inhibitor by the distinctive phenotype of a Δζ knockout mutant in P. denitrificans. ζ blocks exclusively the CCW F1FO-ATPase rotation without affecting the CW-F1FO-ATP synthase turnover, confirming that ζ is important for respiratory bacterial growth by working as a unidirectional pawl-ratchet PdF1FO-ATPase inhibitor, thus preventing the wasteful consumption of cellular ATP. In summary, ζ is a useful model that mimics mitochondrial IF1 but in α-proteobacteria. The structural, functional, and endosymbiotic evolutionary implications of this ζ inhibitor are discussed to shed light on the natural control mechanisms of the three natural inhibitor proteins (ε, ζ, and IF1) of this unique ATP synthase nanomotor, essential for life.


Subject(s)
Adenosine Triphosphate/metabolism , Alphaproteobacteria/enzymology , Enzyme Inhibitors/administration & dosage , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Paracoccus denitrificans/enzymology , Proteins/administration & dosage , Amino Acid Sequence , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Protein Conformation , Protein Subunits , Sequence Homology , ATPase Inhibitory Protein
13.
Biochim Biophys Acta Bioenerg ; 1859(9): 789-796, 2018 09.
Article in English | MEDLINE | ID: mdl-29630891

ABSTRACT

Enzymes in the respiratory chain are increasingly seen as potential targets against multi-drug resistance of human pathogens and cancerous cells. However, a detailed understanding of the mechanism and specificity determinants of known inhibitors is still lacking. Oligomycin, for example, has been known to be an inhibitor of the membrane motor of the mitochondrial ATP synthase for over five decades, and yet little is known about its mode of action at the molecular level. In a recent breakthrough, a crystal structure of the S. cerevisiae c-subunit ring with bound oligomycin revealed the inhibitor docked on the outer face of the proton-binding sites, deep into the transmembrane region. However, the structure of the complex was obtained in an organic solvent rather than detergent or a lipid bilayer, and therefore it has been unclear whether this mode of recognition is physiologically relevant. Here, we use molecular dynamics simulations to address this question and gain insights into the mechanism of oligomycin inhibition. Our findings lead us to propose that oligomycin naturally partitions into the lipid/water interface, and that in this environment the inhibitor can indeed bind to any of the c-ring proton-carrying sites that are exposed to the membrane, thereby becoming an integral component of the proton-coordinating network. As the c-ring rotates within the membrane, driven either by downhill proton permeation or ATP hydrolysis, one of the protonated, oligomycin-bound sites eventually reaches the subunit-a interface and halts the rotary mechanism of the enzyme.


Subject(s)
Adenosine Triphosphate/metabolism , Enzyme Inhibitors/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Oligomycins/metabolism , Saccharomyces cerevisiae/enzymology , Binding Sites , Enzyme Inhibitors/chemistry , Mitochondrial Membranes/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/chemistry , Molecular Dynamics Simulation , Oligomycins/chemistry , Protein Conformation
14.
Carcinogenesis ; 39(9): 1141-1150, 2018 09 21.
Article in English | MEDLINE | ID: mdl-29860383

ABSTRACT

Curcumin has been reported to inhibit inflammation, tumor growth, angiogenesis and metastasis by decreasing cell growth and by inducing apoptosis mainly through the inhibition of nuclear factor kappa-B (NFκB), a master regulator of inflammation. Recent reports also indicate potential metabolic effects of the polyphenol, therefore we analyzed whether and how it affects the energy metabolism of tumor cells. We show that curcumin (10 µM) inhibits the activity of ATP synthase in isolated mitochondrial membranes leading to a dramatic drop of ATP and a reduction of oxygen consumption in in vitro and in vivo tumor models. The effects of curcumin on ATP synthase are independent of the inhibition of NFκB since the IκB Kinase inhibitor, SC-514, does not affect ATP synthase. The activities of the glycolytic enzymes hexokinase, phosphofructokinase, pyruvate kinase and lactate dehydrogenase are only slightly affected in a cell type-specific manner. The energy impairment translates into decreased tumor cell viability. Moreover, curcumin induces apoptosis by promoting the generation of reactive oxygen species (ROS) and malondialdehyde (MDA), a marker of lipid oxidation, and autophagy, at least in part due to the activation of the AMP-activated protein kinase (AMPK). According to the in vitro anti-tumor effect, curcumin (30 mg/kg body weight) significantly delayed in vivo cancer growth likely due to an energy impairment but also through the reduction of tumor angiogenesis. These results establish the ATP synthase, a central enzyme of the cellular energy metabolism, as a target of the antitumoral polyphenol leading to inhibition of cancer cell growth and a general reprogramming of tumor metabolism.


Subject(s)
Antineoplastic Agents/therapeutic use , Curcumin/therapeutic use , Energy Metabolism/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Neoplasms/drug therapy , Oxygen Consumption/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Hexokinase/metabolism , I-kappa B Kinase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Phosphofructokinases/metabolism , Pyruvate Kinase/metabolism , Reactive Oxygen Species/metabolism , Thiophenes/pharmacology
15.
J Biol Chem ; 292(36): 15105-15120, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28673964

ABSTRACT

Sorafenib (Nexavar) is a broad-spectrum multikinase inhibitor that proves effective in treating advanced renal-cell carcinoma and liver cancer. Despite its well-characterized mechanism of action on several established cancer-related protein kinases, sorafenib causes variable responses among human tumors, although the cause for this variation is unknown. In an unbiased screening of an oncology drug library, we found that sorafenib activates recruitment of the ubiquitin E3 ligase Parkin to damaged mitochondria. We show that sorafenib inhibits the activity of both complex II/III of the electron transport chain and ATP synthase. Dual inhibition of these complexes, but not inhibition of each individual complex, stabilizes the serine-threonine protein kinase PINK1 on the mitochondrial outer membrane and activates Parkin. Unlike the protonophore carbonyl cyanide m-chlorophenylhydrazone, which activates the mitophagy response, sorafenib treatment triggers PINK1/Parkin-dependent cellular apoptosis, which is attenuated upon Bcl-2 overexpression. In summary, our results reveal a new mechanism of action for sorafenib as a mitocan and suggest that high Parkin activity levels could make tumor cells more sensitive to sorafenib's actions, providing one possible explanation why Parkin may be a tumor suppressor gene. These insights could be useful in developing new rationally designed combination therapies with sorafenib.


Subject(s)
Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex II/antagonists & inhibitors , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Electron Transport/drug effects , Electron Transport Complex II/metabolism , Electron Transport Complex III/metabolism , HEK293 Cells , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Niacinamide/pharmacology , Sorafenib
16.
J Cell Physiol ; 234(1): 940-949, 2018 01.
Article in English | MEDLINE | ID: mdl-30144054

ABSTRACT

Studies have demonstrated that zebrafish are powerful tools for monitoring environmental toxicity, including radiation hazard. Here we investigated the developmental toxicity of ionizing radiation (IR) in an in vivo embryonic zebrafish model. The effects of heavy ion (12 C6+ ), proton, and X-ray radiation on early zebrafish embryos were determined. A similar dose-dependent decrease in the hatch and survival rate of zebrafish embryos was observed after exposure to these irradiations. Exposure of zebrafish embryos to 1-4 Gy IR caused significant loss of pigmentation. Quantitative real-time reverse transcription polymerase chain reaction, western blot analysis, and in situ hybridization (ISH) experiment revealed that atp5α1 was markedly upregulated in irradiated zebrafish embryos. In addition, IR resulted in a rapid decrease in total adenosine triphosphate (ATP) generation. With dual functions of synthesizing or hydrolyzing ATP, ATP synthase regulated H+ transport crossing the mitochondrial inner. Administration of the mitochondrial ATP synthase inhibitor, oligomycin, partially restored pigmentation in irradiated zebrafish embryos, but the ATPase inhibitor, BTB06584, had no effect. Taken together, these results showed that IR exposure downregulated zebrafish pigmentation through regulation of H+ ion transport in mitochondria.


Subject(s)
Embryonic Development/radiation effects , Pigmentation/radiation effects , Radiation Exposure/adverse effects , Zebrafish/genetics , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Animals , Chlorobenzoates/administration & dosage , DNA Damage/radiation effects , Embryonic Development/genetics , Gene Expression Regulation, Developmental/radiation effects , In Situ Hybridization , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Oligomycins/administration & dosage , Pigmentation/genetics , Radiation, Ionizing , Sulfones/administration & dosage , Zebrafish/growth & development
17.
EMBO J ; 33(7): 762-78, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24521670

ABSTRACT

A key transducer in energy conservation and signaling cell death is the mitochondrial H(+)-ATP synthase. The expression of the ATPase inhibitory factor 1 (IF1) is a strategy used by cancer cells to inhibit the activity of the H(+)-ATP synthase to generate a ROS signal that switches on cellular programs of survival. We have generated a mouse model expressing a mutant of human IF1 in brain neurons to assess the role of the H(+)-ATP synthase in cell death in vivo. The expression of hIF1 inhibits the activity of oxidative phosphorylation and mediates the shift of neurons to an enhanced aerobic glycolysis. Metabolic reprogramming induces brain preconditioning affording protection against quinolinic acid-induced excitotoxicity. Mechanistically, preconditioning involves the activation of the Akt/p70S6K and PARP repair pathways and Bcl-xL protection from cell death. Overall, our findings provide the first in vivo evidence highlighting the H(+)-ATP synthase as a target to prevent neuronal cell death.


Subject(s)
Gene Expression Regulation, Enzymologic , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Proteins/genetics , Signal Transduction , Animals , Apoptosis , Behavior, Animal , Brain/cytology , Brain/drug effects , Brain/enzymology , Glycolysis/drug effects , Humans , Male , Metabolic Networks and Pathways , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Models, Animal , Mutation, Missense , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Neurotoxins/pharmacology , Oxidative Phosphorylation , Promoter Regions, Genetic/genetics , Proteins/metabolism , Quinolinic Acid/pharmacology , Reactive Oxygen Species/metabolism , ATPase Inhibitory Protein
18.
Biol Chem ; 399(2): 197-202, 2018 01 26.
Article in English | MEDLINE | ID: mdl-28976891

ABSTRACT

The mitochondrial F1FO-ATPase is uncompetitively inhibited by NAD+ only when the natural cofactor Mg2+ is replaced by Ca2+, a mode putatively involved in cell death. The Ca2+-dependent F1FO-ATPase is also inhibited when NAD+ concentration in mitochondria is raised by acetoacetate. The enzyme inhibition by NAD+ cannot be ascribed to any de-ac(et)ylation or ADP-ribosylation by sirtuines, as it is not reversed by nicotinamide. Moreover, the addition of acetyl-CoA or palmitate, which would favor the enzyme ac(et)ylation, does not affect the F1FO-ATPase activity. Consistently, NAD+ may play a new role, not associated with redox and non-redox enzymatic reactions, in the Ca2+-dependent regulation of the F1FO-ATPase activity.


Subject(s)
Calcium/metabolism , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , NAD/metabolism , Calcium/pharmacology , Enzyme Activation/drug effects , Humans , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , NAD/pharmacology , Oxidation-Reduction
19.
Mol Vis ; 24: 759-766, 2018.
Article in English | MEDLINE | ID: mdl-30581282

ABSTRACT

Purpose: To identify retinal protein changes that mediate beneficial effects of intravitreal bevacizumab in experimental branch retinal vein occlusion (BRVO). Methods: In six Danish Landrace pigs, BRVO was induced with argon laser in both eyes. After BRVO was induced, the right eye of each animal was given an intravitreal injection of bevacizumab while the left eye was treated with saline water. The retinas were collected 15 days after BRVO, and differentially expressed proteins were analyzed with tandem mass tags-based mass spectrometry. Validation of statistically significantly changed proteins was performed with immunohistochemistry and western blotting. Results: Fluorescein angiography showed no recanalization of the occluded vessels. A total of 4,013 proteins were successfully identified and quantified. Nine proteins were statistically significantly changed following bevacizumab intervention. In experimental BRVO, bevacizumab treatment resulted in upregulation of transthyretin (TTR) and pantothenate kinase 3. Bevacizumab downregulated protocadherin 7, protein FAM192A, and ATP synthase protein 8. Immunohistochemistry revealed that TTR was highly abundant in the choroid following bevacizumab intervention. Conclusions: Bevacizumab intervention in experimental BRVO resulted in an increased level of TTR. This is the second study in which we showed an increased retinal level of TTR following anti-vascular endothelial growth factor (VEGF) intervention in experimental BRVO. We hypothesize that there is an interaction between TTR and VEGF and that bevacizumab may exert a beneficial effect on the retina by upregulating TTR.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Gene Expression Regulation/drug effects , Prealbumin/genetics , Retina/drug effects , Retinal Vein Occlusion/drug therapy , Animals , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Choroid/blood supply , Choroid/diagnostic imaging , Choroid/drug effects , Choroid/metabolism , Fluorescein Angiography , Gene Expression Profiling , Humans , Immunoglobulin gamma-Chains/genetics , Immunoglobulin gamma-Chains/metabolism , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/metabolism , Intravitreal Injections , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prealbumin/agonists , Prealbumin/metabolism , Retina/diagnostic imaging , Retina/metabolism , Retina/pathology , Retinal Vein Occlusion/diagnostic imaging , Retinal Vein Occlusion/genetics , Retinal Vein Occlusion/pathology , Swine
20.
Proc Natl Acad Sci U S A ; 112(6): 1850-5, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25624475

ABSTRACT

Flagella are multiprotein complexes necessary for swimming and swarming motility. In Salmonella enterica serovar Typhimurium, flagella-mediated motility is repressed by the PhoP/PhoQ regulatory system. We now report that Salmonella can move on 0.3% agarose media in a flagella-independent manner when experiencing the PhoP/PhoQ-inducing signal low Mg(2+). This motility requires the PhoP-activated mgtA, mgtC, and pagM genes, which specify a Mg(2+) transporter, an inhibitor of Salmonella's own F1Fo ATPase, and a small protein of unknown function, respectively. The MgtA and MgtC proteins are necessary for pagM expression because pagM mRNA levels were lower in mgtA and mgtC mutants than in wild-type Salmonella, and also because pagM expression from a heterologous promoter rescued motility in mgtA and mgtC mutants. PagM promotes group motility by a surface protein(s), as a pagM-expressing strain conferred motility upon a pagM null mutant, and proteinase K treatment eliminated motility. The pagM gene is rarely found outside subspecies I of S. enterica and often present in nonfunctional allelic forms in organisms lacking the identified motility. Deletion of the pagM gene reduced bacterial replication on 0.3% agarose low Mg(2+) media but not in low Mg(2+) liquid media. Our findings define a form of motility that allows Salmonella to scavenge nutrients and to escape toxic compounds in low Mg(2+) semisolid environments.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Cation Transport Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Membrane Transport Proteins/metabolism , Movement/physiology , Salmonella typhimurium/physiology , Amino Acid Sequence , Base Sequence , Computational Biology , Flagella/metabolism , Magnesium/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Molecular Sequence Data , Mutagenesis , Sequence Alignment , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL