Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 133(1)2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36301669

RESUMEN

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.


Asunto(s)
Cobre , Enfermedades Mitocondriales , Ratones , Animales , Cobre/metabolismo , alfa-Fetoproteínas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Terapia de Inmunosupresión
2.
Sci Rep ; 12(1): 15663, 2022 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-36123435

RESUMEN

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.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Adenosina Trifosfatasas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cladribina/uso terapéutico , Doxorrubicina/uso terapéutico , Excipientes , Homoharringtonina/farmacología , Humanos , Nucleósidos/uso terapéutico , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Neoplasias de la Mama Triple Negativas/patología
3.
Cell Rep ; 36(11): 109704, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525369

RESUMEN

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.


Asunto(s)
Núcleo Celular/metabolismo , Histonas/genética , Mitocondrias/metabolismo , Transcriptoma , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Histonas/deficiencia , Histonas/metabolismo , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Proteínas Mitocondriales/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Fosforilación Oxidativa , Células de Purkinje/citología , Células de Purkinje/metabolismo , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba
4.
Elife ; 102021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33591272

RESUMEN

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.


Asunto(s)
Evolución Biológica , Proteínas Mitocondriales/genética , Proteínas de Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas Transportadoras de Cobre/genética , Proteínas Transportadoras de Cobre/metabolismo , Eucariontes , Ratones , Mitocondrias , Proteínas Mitocondriales/metabolismo , Mutagénesis Sitio-Dirigida , Proteínas de Transporte de Fosfato/genética , Proteínas de Transporte de Fosfato/metabolismo , Filogenia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
5.
Cell Rep ; 29(12): 4114-4126.e5, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851937

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Proteínas Mitocondriales/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Proteínas Portadoras/genética , Complejo IV de Transporte de Electrones/genética , Humanos , Espectroscopía de Resonancia Magnética , Proteínas Mitocondriales/genética , Chaperonas Moleculares/metabolismo , Mutación/genética , Unión Proteica , Proteína Disulfuro Reductasa (Glutatión)/genética
6.
Proc Natl Acad Sci U S A ; 115(32): 8161-8166, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30038027

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Cobre/deficiencia , Drogas en Investigación/farmacología , Complejo IV de Transporte de Electrones/metabolismo , Hidrazinas/farmacología , Mitocondrias/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Transporte Biológico/genética , Proteínas Portadoras/genética , Línea Celular , Coenzimas/deficiencia , Cobre/uso terapéutico , Transportador de Cobre 1 , Suplementos Dietéticos , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Drogas en Investigación/uso terapéutico , Fibroblastos , Humanos , Hidrazinas/uso terapéutico , Proteínas de Transporte de Membrana/genética , Errores Innatos del Metabolismo/tratamiento farmacológico , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Chaperonas Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Ratas , Saccharomyces cerevisiae , Pez Cebra , Proteínas de Pez Cebra/genética
7.
J Biol Chem ; 293(6): 1887-1896, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29237729

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Transporte de Fosfato/metabolismo , Proteínas Transportadoras de Solutos/metabolismo , Animales , Transporte Biológico , Proteínas de Transporte de Catión/genética , Complejo IV de Transporte de Electrones/genética , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas de Transporte de Fosfato/genética , Proteínas Transportadoras de Solutos/genética
8.
Hum Mol Genet ; 26(23): 4617-4628, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973536

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Membrana Celular/metabolismo , Cobre/deficiencia , Transportador de Cobre 1 , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/genética , Homeostasis , Transporte Iónico , Metalochaperonas/genética , Metalochaperonas/metabolismo , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Transducción de Señal
9.
Cell Rep ; 10(6): 933-943, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25683716

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-24403053

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclooxigenasa 2/metabolismo , Deficiencia de Citocromo-c Oxidasa/enzimología , Complejo IV de Transporte de Electrones/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Portadoras/genética , Línea Celular , Ciclooxigenasa 2/genética , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Humanos , Proteínas de la Membrana/genética , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Chaperonas Moleculares , Estabilidad Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...