Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(1)2023 01 03.
Article in English | MEDLINE | ID: mdl-36301669

ABSTRACT

Signaling circuits crucial to systemic physiology are widespread, yet uncovering their molecular underpinnings remains a barrier to understanding the etiology of many metabolic disorders. Here, we identified a copper-linked signaling circuit activated by disruption of mitochondrial function in the murine liver or heart that resulted in atrophy of the spleen and thymus and caused a peripheral white blood cell deficiency. We demonstrated that the leukopenia was caused by α-fetoprotein, which required copper and the cell surface receptor CCR5 to promote white blood cell death. We further showed that α-fetoprotein expression was upregulated in several cell types upon inhibition of oxidative phosphorylation. Collectively, our data argue that α-fetoprotein may be secreted by bioenergetically stressed tissue to suppress the immune system, an effect that may explain the recurrent or chronic infections that are observed in a subset of mitochondrial diseases or in other disorders with secondary mitochondrial dysfunction.


Subject(s)
Copper , Mitochondrial Diseases , Mice , Animals , Copper/metabolism , alpha-Fetoproteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Immunosuppression Therapy
2.
Sci Rep ; 12(1): 15663, 2022 09 19.
Article in English | MEDLINE | ID: mdl-36123435

ABSTRACT

The lack of targeted therapies for triple-negative breast cancer (TNBC) contributes to their high mortality rates and high risk of relapse compared to other subtypes of breast cancer. Most TNBCs (75%) have downregulated the expression of CREB3L1 (cAMP-responsive element binding protein 3 like 1), a transcription factor and metastasis suppressor that represses genes that promote cancer progression and metastasis. In this report, we screened an FDA-approved drug library and identified four drugs that were highly cytotoxic towards HCC1806 CREB3L1-deficient TNBC cells. These four drugs were: (1) palbociclib isethionate, a CDK4/6 inhibitor, (2) lanatocide C (also named isolanid), a Na+/K+-ATPase inhibitor, (3) cladribine, a nucleoside analog, and (4) homoharringtonine (also named omacetaxine mepesuccinate), a protein translation inhibitor. Homoharringtonine consistently showed the most cytotoxicity towards an additional six TNBC cell lines (BT549, HCC1395, HCC38, Hs578T, MDA-MB-157, MDA-MB-436), and several luminal A breast cancer cell lines (HCC1428, MCF7, T47D, ZR-75-1). All four drugs were then separately evaluated for possible synergy with the chemotherapy agents, doxorubicin (an anthracycline) and paclitaxel (a microtubule stabilizing agent). A strong synergy was observed using the combination of homoharringtonine and paclitaxel, with high cytotoxicity towards TNBC cells at lower concentrations than when each was used separately.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Adenosine Triphosphatases , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cladribine/therapeutic use , Doxorubicin/therapeutic use , Excipients , Homoharringtonine/pharmacology , Humans , Nucleosides/therapeutic use , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Triple Negative Breast Neoplasms/pathology
3.
Cell Rep ; 36(11): 109704, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525369

ABSTRACT

Histone variants are crucial regulators of chromatin structure and gene transcription, yet their functions within the brain remain largely unexplored. Here, we show that the H2A histone variant H2A.Z is essential for neuronal survival. Mice lacking H2A.Z in GABAergic neurons or Purkinje cells (PCs) present with a progressive cerebellar ataxia accompanied by widespread degeneration of PCs. Ablation of H2A.Z in other neuronal subtypes also triggers cell death. H2A.Z binds to the promoters of key nuclear-encoded mitochondrial genes to regulate their expression and promote organelle function. Bolstering mitochondrial activity genetically or by organelle transplant enhances the survival of H2A.Z-ablated neurons. Changes in bioenergetic status alter H2A.Z occupancy at the promoters of nuclear-encoded mitochondrial genes, an adaptive response essential for cell survival. Our results highlight that H2A.Z fulfills a key, conserved role in neuronal survival by acting as a transcriptional rheostat to regulate the expression of genes critical to mitochondrial function.


Subject(s)
Cell Nucleus/metabolism , Histones/genetics , Mitochondria/metabolism , Transcriptome , Animals , Cell Survival/drug effects , Cells, Cultured , Down-Regulation , Fibroblasts/cytology , Fibroblasts/metabolism , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Histones/deficiency , Histones/metabolism , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Phosphorylation , Purkinje Cells/cytology , Purkinje Cells/metabolism , Transcriptome/drug effects , Up-Regulation
4.
Elife ; 102021 02 16.
Article in English | MEDLINE | ID: mdl-33591272

ABSTRACT

The mitochondrial carrier family protein SLC25A3 transports both copper and phosphate in mammals, yet in Saccharomyces cerevisiae the transport of these substrates is partitioned across two paralogs: PIC2 and MIR1. To understand the ancestral state of copper and phosphate transport in mitochondria, we explored the evolutionary relationships of PIC2 and MIR1 orthologs across the eukaryotic tree of life. Phylogenetic analyses revealed that PIC2-like and MIR1-like orthologs are present in all major eukaryotic supergroups, indicating an ancient gene duplication created these paralogs. To link this phylogenetic signal to protein function, we used structural modeling and site-directed mutagenesis to identify residues involved in copper and phosphate transport. Based on these analyses, we generated an L175A variant of mouse SLC25A3 that retains the ability to transport copper but not phosphate. This work highlights the utility of using an evolutionary framework to uncover amino acids involved in substrate recognition by mitochondrial carrier family proteins.


Subject(s)
Biological Evolution , Mitochondrial Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Eukaryota , Mice , Mitochondria , Mitochondrial Proteins/metabolism , Mutagenesis, Site-Directed , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , Phylogeny , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
5.
Cell Rep ; 29(12): 4114-4126.e5, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851937

ABSTRACT

In eukaryotes, cellular respiration is driven by mitochondrial cytochrome c oxidase (CcO), an enzyme complex that requires copper cofactors for its catalytic activity. Insertion of copper into its catalytically active subunits, including COX2, is a complex process that requires metallochaperones and redox proteins including SCO1, SCO2, and COA6, a recently discovered protein whose molecular function is unknown. To uncover the molecular mechanism by which COA6 and SCO proteins mediate copper delivery to COX2, we have solved the solution structure of COA6, which reveals a coiled-coil-helix-coiled-coil-helix domain typical of redox-active proteins found in the mitochondrial inter-membrane space. Accordingly, we demonstrate that COA6 can reduce the copper-coordinating disulfides of its client proteins, SCO1 and COX2, allowing for copper binding. Finally, our determination of the interaction surfaces and reduction potentials of COA6 and its client proteins provides a mechanism of how metallochaperone and disulfide reductase activities are coordinated to deliver copper to CcO.


Subject(s)
Carrier Proteins/metabolism , Electron Transport Complex IV/metabolism , Mitochondrial Proteins/metabolism , Protein Disulfide Reductase (Glutathione)/metabolism , Carrier Proteins/genetics , Electron Transport Complex IV/genetics , Humans , Magnetic Resonance Spectroscopy , Mitochondrial Proteins/genetics , Molecular Chaperones/metabolism , Mutation/genetics , Protein Binding , Protein Disulfide Reductase (Glutathione)/genetics
6.
Proc Natl Acad Sci U S A ; 115(32): 8161-8166, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30038027

ABSTRACT

Copper is an essential cofactor of cytochrome c oxidase (CcO), the terminal enzyme of the mitochondrial respiratory chain. Inherited loss-of-function mutations in several genes encoding proteins required for copper delivery to CcO result in diminished CcO activity and severe pathologic conditions in affected infants. Copper supplementation restores CcO function in patient cells with mutations in two of these genes, COA6 and SCO2, suggesting a potential therapeutic approach. However, direct copper supplementation has not been therapeutically effective in human patients, underscoring the need to identify highly efficient copper transporting pharmacological agents. By using a candidate-based approach, we identified an investigational anticancer drug, elesclomol (ES), that rescues respiratory defects of COA6-deficient yeast cells by increasing mitochondrial copper content and restoring CcO activity. ES also rescues respiratory defects in other yeast mutants of copper metabolism, suggesting a broader applicability. Low nanomolar concentrations of ES reinstate copper-containing subunits of CcO in a zebrafish model of copper deficiency and in a series of copper-deficient mammalian cells, including those derived from a patient with SCO2 mutations. These findings reveal that ES can restore intracellular copper homeostasis by mimicking the function of missing transporters and chaperones of copper, and may have potential in treating human disorders of copper metabolism.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/deficiency , Drugs, Investigational/pharmacology , Electron Transport Complex IV/metabolism , Hydrazines/pharmacology , Mitochondria/drug effects , Animals , Antineoplastic Agents/therapeutic use , Biological Transport/genetics , Carrier Proteins/genetics , Cell Line , Coenzymes/deficiency , Copper/therapeutic use , Copper Transporter 1 , Dietary Supplements , Disease Models, Animal , Drug Repositioning , Drugs, Investigational/therapeutic use , Fibroblasts , Humans , Hydrazines/therapeutic use , Membrane Transport Proteins/genetics , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molecular Chaperones , Mutagenesis, Site-Directed , Mutation , Rats , Saccharomyces cerevisiae , Zebrafish , Zebrafish Proteins/genetics
7.
J Biol Chem ; 293(6): 1887-1896, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29237729

ABSTRACT

Copper is required for the activity of cytochrome c oxidase (COX), the terminal electron-accepting complex of the mitochondrial respiratory chain. The likely source of copper used for COX biogenesis is a labile pool found in the mitochondrial matrix. In mammals, the proteins that transport copper across the inner mitochondrial membrane remain unknown. We previously reported that the mitochondrial carrier family protein Pic2 in budding yeast is a copper importer. The closest Pic2 ortholog in mammalian cells is the mitochondrial phosphate carrier SLC25A3. Here, to investigate whether SLC25A3 also transports copper, we manipulated its expression in several murine and human cell lines. SLC25A3 knockdown or deletion consistently resulted in an isolated COX deficiency in these cells, and copper addition to the culture medium suppressed these biochemical defects. Consistent with a conserved role for SLC25A3 in copper transport, its heterologous expression in yeast complemented copper-specific defects observed upon deletion of PIC2 Additionally, assays in Lactococcus lactis and in reconstituted liposomes directly demonstrated that SLC25A3 functions as a copper transporter. Taken together, these data indicate that SLC25A3 can transport copper both in vitro and in vivo.


Subject(s)
Cation Transport Proteins/metabolism , Copper/metabolism , Electron Transport Complex IV/metabolism , Mitochondrial Proteins/metabolism , Phosphate Transport Proteins/metabolism , Solute Carrier Proteins/metabolism , Animals , Biological Transport , Cation Transport Proteins/genetics , Electron Transport Complex IV/genetics , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Phosphate Transport Proteins/genetics , Solute Carrier Proteins/genetics
8.
Hum Mol Genet ; 26(23): 4617-4628, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973536

ABSTRACT

SCO1 is a ubiquitously expressed, mitochondrial protein with essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper homeostasis. SCO1 patients present with severe forms of early onset disease, and ultimately succumb from liver, heart or brain failure. However, the inherent susceptibility of these tissues to SCO1 mutations and the clinical heterogeneity observed across SCO1 pedigrees remain poorly understood phenomena. To further address this issue, we generated Sco1hrt/hrt and Sco1stm/stm mice in which Sco1 was specifically deleted in heart and striated muscle, respectively. Lethality was observed in both models due to a combined COX and copper deficiency that resulted in a dilated cardiomyopathy. Left ventricular dilation and loss of heart function was preceded by a temporal decrease in COX activity and copper levels in the longer-lived Sco1stm/stm mice. Interestingly, the reduction in copper content of Sco1stm/stm cardiomyocytes was due to the mislocalisation of CTR1, the high affinity transporter that imports copper into the cell. CTR1 was similarly mislocalized to the cytosol in the heart of knockin mice carrying a homozygous G115S substitution in Sco1, which in humans causes a hypertrophic cardiomyopathy. Our current findings in the heart are in marked contrast to our prior observations in the liver, where Sco1 deletion results in a near complete absence of CTR1 protein. These data collectively argue that mutations perturbing SCO1 function have tissue-specific consequences for the machinery that ultimately governs copper homeostasis, and further establish the importance of aberrant mitochondrial signaling to the etiology of copper handling disorders.


Subject(s)
Cation Transport Proteins/metabolism , Copper/metabolism , Electron Transport Complex IV/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Animals , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cell Membrane/metabolism , Copper/deficiency , Copper Transporter 1 , Disease Models, Animal , Electron Transport Complex IV/genetics , Homeostasis , Ion Transport , Metallochaperones/genetics , Metallochaperones/metabolism , Mice , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Signal Transduction
9.
Cell Rep ; 10(6): 933-943, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25683716

ABSTRACT

Human SCO1 fulfills essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper (Cu) homeostasis, yet it remains unclear why pathogenic mutations in this gene cause such clinically heterogeneous forms of disease. Here, we establish a Sco1 mouse model of human disease and show that ablation of Sco1 expression in the liver is lethal owing to severe COX and Cu deficiencies. We further demonstrate that the Cu deficiency is explained by a functional connection between SCO1 and CTR1, the high-affinity transporter that imports Cu into the cell. CTR1 is rapidly degraded in the absence of SCO1 protein, and we show that its levels are restored in Sco1-/- mouse embryonic fibroblasts upon inhibition of the proteasome. These data suggest that mitochondrial signaling through SCO1 provides a post-translational mechanism to regulate CTR1-dependent Cu import into the cell, and they further underpin the importance of mitochondria in cellular Cu homeostasis.

10.
Hum Mol Genet ; 23(11): 2901-13, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24403053

ABSTRACT

Cytochrome c oxidase (CIV) deficiency is one of the most common respiratory chain defects in patients presenting with mitochondrial encephalocardiomyopathies. CIV biogenesis is complicated by the dual genetic origin of its structural subunits, and assembly of a functional holoenzyme complex requires a large number of nucleus-encoded assembly factors. In general, the functions of these assembly factors remain poorly understood, and mechanistic investigations of human CIV biogenesis have been limited by the availability of model cell lines. Here, we have used small interference RNA and transcription activator-like effector nucleases (TALENs) technology to create knockdown and knockout human cell lines, respectively, to study the function of the CIV assembly factor COX20 (FAM36A). These cell lines exhibit a severe, isolated CIV deficiency due to instability of COX2, a mitochondrion-encoded CIV subunit. Mitochondria lacking COX20 accumulate CIV subassemblies containing COX1 and COX4, similar to those detected in fibroblasts from patients carrying mutations in the COX2 copper chaperones SCO1 and SCO2. These results imply that in the absence of COX20, COX2 is inefficiently incorporated into early CIV subassemblies. Immunoprecipitation assays using a stable COX20 knockout cell line expressing functional COX20-FLAG allowed us to identify an interaction between COX20 and newly synthesized COX2. Additionally, we show that SCO1 and SCO2 act on COX20-bound COX2. We propose that COX20 acts as a chaperone in the early steps of COX2 maturation, stabilizing the newly synthesized protein and presenting COX2 to its metallochaperone module, which in turn facilitates the incorporation of mature COX2 into the CIV assembly line.


Subject(s)
Carrier Proteins/metabolism , Cyclooxygenase 2/metabolism , Cytochrome-c Oxidase Deficiency/enzymology , Electron Transport Complex IV/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Carrier Proteins/genetics , Cell Line , Cyclooxygenase 2/genetics , Cytochrome-c Oxidase Deficiency/genetics , Cytochrome-c Oxidase Deficiency/metabolism , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/genetics , Humans , Membrane Proteins/genetics , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molecular Chaperones , Protein Stability
SELECTION OF CITATIONS
SEARCH DETAIL
...