Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(3): 101719, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35151690

RESUMEN

The mitochondrial protein LonP1 is an ATP-dependent protease that mitigates cell stress and calibrates mitochondrial metabolism and energetics. Biallelic mutations in the LONP1 gene are known to cause a broad spectrum of diseases, and LonP1 dysregulation is also implicated in cancer and age-related disorders. Despite the importance of LonP1 in health and disease, specific inhibitors of this protease are unknown. Here, we demonstrate that 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO) and its -methyl and -imidazole derivatives reversibly inhibit LonP1 by a noncompetitive mechanism, blocking ATP-hydrolysis and thus proteolysis. By contrast, we found that CDDO-anhydride inhibits the LonP1 ATPase competitively. Docking of CDDO derivatives in the cryo-EM structure of LonP1 shows these compounds bind a hydrophobic pocket adjacent to the ATP-binding site. The binding site of CDDO derivatives was validated by amino acid substitutions that increased LonP1 inhibition and also by a pathogenic mutation that causes cerebral, ocular, dental, auricular and skeletal (CODAS) syndrome, which ablated inhibition. CDDO failed to inhibit the ATPase activity of the purified 26S proteasome, which like LonP1 belongs to the AAA+ superfamily of ATPases Associated with diverse cellular Activities, suggesting that CDDO shows selectivity within this family of ATPases. Furthermore, we show that noncytotoxic concentrations of CDDO derivatives in cultured cells inhibited LonP1, but not the 26S proteasome. Taken together, these findings provide insights for future development of LonP1-specific inhibitors with chemotherapeutic potential.


Asunto(s)
Proteasas ATP-Dependientes , Adenosina Trifosfato , Mitocondrias , Proteínas Mitocondriales , Ácido Oleanólico/análogos & derivados , Adenosina Trifosfato/metabolismo , Endopeptidasas/metabolismo , Hidrólisis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Ácido Oleanólico/farmacología
2.
Hum Mol Genet ; 28(2): 290-306, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30304514

RESUMEN

LonP1 is crucial for maintaining mitochondrial proteostasis and mitigating cell stress. We identified a novel homozygous missense LONP1 variant, c.2282 C > T, (p.Pro761Leu), by whole-exome and Sanger sequencing in two siblings born to healthy consanguineous parents. Both siblings presented with stepwise regression during infancy, profound hypotonia and muscle weakness, severe intellectual disability and progressive cerebellar atrophy on brain imaging. Muscle biopsy revealed the absence of ragged-red fibers, however, scattered cytochrome c oxidase-negative staining and electron dense mitochondrial inclusions were observed. Primary cultured fibroblasts from the siblings showed normal levels of mtDNA and mitochondrial transcripts, and normal activities of oxidative phosphorylation complexes I through V. Interestingly, fibroblasts of both siblings showed glucose-repressed oxygen consumption compared to their mother, whereas galactose and palmitic acid utilization were similar. Notably, the siblings' fibroblasts had reduced pyruvate dehydrogenase (PDH) activity and elevated intracellular lactate:pyruvate ratios, whereas plasma ratios were normal. We demonstrated that in the siblings' fibroblasts, PDH dysfunction was caused by increased levels of the phosphorylated E1α subunit of PDH, which inhibits enzyme activity. Blocking E1α phosphorylation activated PDH and reduced intracellular lactate concentrations. In addition, overexpressing wild-type LonP1 in the siblings' fibroblasts down-regulated phosphoE1α. Furthermore, in vitro studies demonstrated that purified LonP1-P761L failed to degrade phosphorylated E1α, in contrast to wild-type LonP1. We propose a novel mechanism whereby homozygous expression of the LonP1-P761L variant leads to PDH deficiency and energy metabolism dysfunction, which promotes severe neurologic impairment and neurodegeneration.


Asunto(s)
Proteasas ATP-Dependientes/genética , Enfermedades Cerebelosas/genética , Proteínas Mitocondriales/genética , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/genética , Alelos , Enfermedades Cerebelosas/enzimología , ADN Mitocondrial/metabolismo , Homocigoto , Humanos , Recién Nacido , Lactatos/metabolismo , Masculino , Enfermedades Neurodegenerativas/enzimología , Linaje , Fosforilación , Subunidades de Proteína/metabolismo , Proteolisis , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/patología
3.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R547-R562, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33112656

RESUMEN

Mitochondria play key roles in the differentiation and maturation of human cardiomyocytes (CMs). As human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold potential in the treatment of heart diseases, we sought to identify key mitochondrial pathways and regulators, which may provide targets for improving cardiac differentiation and maturation. Proteomic analysis was performed on enriched mitochondrial protein extracts isolated from hiPSC-CMs differentiated from dermal fibroblasts (dFCM) and cardiac fibroblasts (cFCM) at time points between 12 and 115 days of differentiation, and from adult and neonatal mouse hearts. Mitochondrial proteins with a twofold change at time points up to 120 days relative to 12 days were subjected to ingenuity pathway analysis (IPA). The highest upregulation was in metabolic pathways for fatty acid oxidation (FAO), the tricarboxylic acid (TCA) cycle, oxidative phosphorylation (OXPHOS), and branched chain amino acid (BCAA) degradation. The top upstream regulators predicted to be activated were peroxisome proliferator-activated receptor γ coactivator 1 α (PGC1-α), the insulin receptor (IR), and the retinoblastoma protein (Rb1) transcriptional repressor. IPA and immunoblotting showed upregulation of the mitochondrial LonP1 protease-a regulator of mitochondrial proteostasis, energetics, and metabolism. LonP1 knockdown increased FAO in neonatal rat ventricular cardiomyocytes (nRVMs). Our results support the notion that LonP1 upregulation negatively regulates FAO in cardiomyocytes to calibrate the flux between glucose and fatty acid oxidation. We discuss potential mechanisms by which IR, Rb1, and LonP1 regulate the metabolic shift from glycolysis to OXPHOS and FAO. These newly identified factors and pathways may help in optimizing the maturation of iPSC-CMs.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/metabolismo , Biogénesis de Organelos , Proteoma , Proteómica , Animales , Línea Celular , Linaje de la Célula , Metabolismo Energético , Humanos , Ratones , Mitocondrias Cardíacas/genética , Proteínas Mitocondriales/genética , Ratas , Factores de Tiempo
4.
Mol Cell ; 49(1): 121-32, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23201127

RESUMEN

Human mitochondrial transcription factor A (TFAM) is a high-mobility group (HMG) protein at the nexus of mitochondrial DNA (mtDNA) replication, transcription, and inheritance. Little is known about the mechanisms underlying its posttranslational regulation. Here, we demonstrate that TFAM is phosphorylated within its HMG box 1 (HMG1) by cAMP-dependent protein kinase in mitochondria. HMG1 phosphorylation impairs the ability of TFAM to bind DNA and to activate transcription. We show that only DNA-free TFAM is degraded by the Lon protease, which is inhibited by the anticancer drug bortezomib. In cells with normal mtDNA levels, HMG1-phosphorylated TFAM is degraded by Lon. However, in cells with severe mtDNA deficits, nonphosphorylated TFAM is also degraded, as it is DNA free. Depleting Lon in these cells increases levels of TFAM and upregulates mtDNA content, albeit transiently. Phosphorylation and proteolysis thus provide mechanisms for rapid fine-tuning of TFAM function and abundance in mitochondria, which are crucial for maintaining and expressing mtDNA.


Asunto(s)
ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteasa La/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Transcripción/metabolismo , Sustitución de Aminoácidos , Secuencia de Bases , Sitios de Unión , Ácidos Borónicos/farmacología , Bortezomib , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Genoma Mitocondrial , Células HEK293 , Células HeLa , Humanos , Mitocondrias/enzimología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Modelos Moleculares , Fosforilación , Proteasa La/antagonistas & inhibidores , Proteasa La/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteolisis , Pirazinas/farmacología , Interferencia de ARN , Factores de Transcripción/química , Factores de Transcripción/genética , Activación Transcripcional
5.
Nucleic Acids Res ; 46(7): 3633-3642, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29897602

RESUMEN

Mitochondrial transcription factor A (TFAM) is essential for the maintenance, expression and transmission of mitochondrial DNA (mtDNA). However, mechanisms for the post-translational regulation of TFAM are poorly understood. Here, we show that TFAM is lysine acetylated within its high-mobility-group box 1, a domain that can also be serine phosphorylated. Using bulk and single-molecule methods, we demonstrate that site-specific phosphoserine and acetyl-lysine mimics of human TFAM regulate its interaction with non-specific DNA through distinct kinetic pathways. We show that higher protein concentrations of both TFAM mimics are required to compact DNA to a similar extent as the wild-type. Compaction is thought to be crucial for regulating mtDNA segregation and expression. Moreover, we reveal that the reduced DNA binding affinity of the acetyl-lysine mimic arises from a lower on-rate, whereas the phosphoserine mimic displays both a decreased on-rate and an increased off-rate. Strikingly, the increased off-rate of the phosphoserine mimic is coupled to a significantly faster diffusion of TFAM on DNA. These findings indicate that acetylation and phosphorylation of TFAM can fine-tune TFAM-DNA binding affinity, to permit the discrete regulation of mtDNA dynamics. Furthermore, our results suggest that phosphorylation could additionally regulate transcription by altering the ability of TFAM to locate promoter sites.


Asunto(s)
ADN Mitocondrial/genética , Proteínas de Unión al ADN/genética , Proteínas Mitocondriales/genética , Factores de Transcripción/genética , Transcripción Genética , Acetilación , ADN Mitocondrial/química , Proteínas de Unión al ADN/química , Humanos , Cinética , Proteínas Mitocondriales/química , Fosforilación , Regiones Promotoras Genéticas , Factores de Transcripción/química
6.
J Mol Cell Cardiol ; 128: 38-50, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30625302

RESUMEN

RATIONALE: LonP1 is an essential mitochondrial protease, which is crucial for maintaining mitochondrial proteostasis and mitigating cell stress. However, the importance of LonP1 during cardiac stress is largely unknown. OBJECTIVE: To determine the functions of LonP1 during ischemia/reperfusion (I/R) injury in vivo, and hypoxia-reoxygenation (H/R) stress in vitro. METHODS AND RESULTS: LonP1 was induced 2-fold in wild-type mice during cardiac ischemic preconditioning (IPC), which protected the heart against ischemia-reperfusion (I/R) injury. In contrast, haploinsufficiency of LonP1 (LONP1+/-) abrogated IPC-mediated cardioprotection. Furthermore, LONP1+/- mice showed significantly increased infarct size after I/R injury, whereas mice with 3-4 fold cardiac-specific overexpression of LonP1 (LonTg) had substantially smaller infarct size and reduced apoptosis compared to wild-type controls. To investigate the mechanisms underlying cardioprotection, LonTg mice were subjected to ischemia (45 min) followed by short intervals of reperfusion (10, 30, 120 min). During early reperfusion, the left ventricles of LonTg mice showed substantially reduced oxidative protein damage, maintained mitochondrial redox homeostasis, and showed a marked downregulation of both Complex I protein level and activity in contrast to NTg mice. Conversely, when LonP1 was knocked down in isolated neonatal rat ventricular myocytes (NRVMs), an up-regulation of Complex I subunits and electron transport chain (ETC) activities was observed, which was associated with increased superoxide production and reduced respiratory efficiency. The knockdown of LonP1 in NRVMs caused a striking dysmorphology of the mitochondrial inner membrane, mitochondrial hyperpolarization and increased hypoxia-reoxygenation (H/R)-activated apoptosis. Whereas, LonP1 overexpression blocked H/R-induced cell death. CONCLUSIONS: LonP1 is an endogenous mediator of cardioprotection. Our findings show that upregulation of LonP1 mitigates cardiac injury by preventing oxidative damage of proteins and lipids, preserving mitochondrial redox balance and reprogramming bioenergetics by reducing Complex I content and activity. Mechanisms that promote the upregulation of LonP1 could be beneficial in protecting the myocardium from cardiac stress and limiting I/R injury.


Asunto(s)
Proteasas ATP-Dependientes/genética , Proteínas Mitocondriales/genética , Infarto del Miocardio/genética , Estrés Oxidativo/genética , Daño por Reperfusión/genética , Animales , Animales Recién Nacidos , Apoptosis/genética , Complejo I de Transporte de Electrón/genética , Regulación de la Expresión Génica/genética , Precondicionamiento Isquémico Miocárdico , Lípidos/genética , Ratones , Mitocondrias/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Especies Reactivas de Oxígeno , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Superóxidos/metabolismo
7.
Bioorg Chem ; 88: 102913, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31015177

RESUMEN

A new series of ß-Carboline/Schiff bases was designed, synthesized, characterised and biologically evaluated as inhibitors of PLK-1. The synthesized compounds exhibited strong to moderate cytotoxic activities against NCI-60 panel cell assay. Compound SB-2 was the most potent, particularly against colon with GI50 of 3-45 µM on NCI-60 panel cell lines. SB-2 selectively inhibited PLK-1 at 15 µM on KinomeScan screening. It also showed a dose-dependent cell cycle arrest at S/G2 phase on HCT-116 and induced apoptosis by the activation of procaspase-3 and cleaved PARP. Further, the antitumor studies on DLA and EAC model revealed that SB-2, at 100 mg/kg/bd.wt significantly increased their average lifespan. Further, a decrease in the body weight of the tumor-bearing mice was also observed when compared to the tumor controlled mice. SB-2 thus shows good potential as antitumor agent.


Asunto(s)
Antineoplásicos/uso terapéutico , Carbolinas/uso terapéutico , Carcinoma de Ehrlich/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Carbolinas/síntesis química , Carbolinas/farmacocinética , Carbolinas/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/toxicidad , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Bibliotecas de Moléculas Pequeñas/toxicidad , Células Vero
8.
Am J Hum Genet ; 96(1): 121-35, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25574826

RESUMEN

CODAS syndrome is a multi-system developmental disorder characterized by cerebral, ocular, dental, auricular, and skeletal anomalies. Using whole-exome and Sanger sequencing, we identified four LONP1 mutations inherited as homozygous or compound-heterozygous combinations among ten individuals with CODAS syndrome. The individuals come from three different ancestral backgrounds (Amish-Swiss from United States, n = 8; Mennonite-German from Canada, n = 1; mixed European from Canada, n = 1). LONP1 encodes Lon protease, a homohexameric enzyme that mediates protein quality control, respiratory-complex assembly, gene expression, and stress responses in mitochondria. All four pathogenic amino acid substitutions cluster within the AAA(+) domain at residues near the ATP-binding pocket. In biochemical assays, pathogenic Lon proteins show substrate-specific defects in ATP-dependent proteolysis. When expressed recombinantly in cells, all altered Lon proteins localize to mitochondria. The Old Order Amish Lon variant (LONP1 c.2161C>G[p.Arg721Gly]) homo-oligomerizes poorly in vitro. Lymphoblastoid cell lines generated from affected children have (1) swollen mitochondria with electron-dense inclusions and abnormal inner-membrane morphology; (2) aggregated MT-CO2, the mtDNA-encoded subunit II of cytochrome c oxidase; and (3) reduced spare respiratory capacity, leading to impaired mitochondrial proteostasis and function. CODAS syndrome is a distinct, autosomal-recessive, developmental disorder associated with dysfunction of the mitochondrial Lon protease.


Asunto(s)
Proteasas ATP-Dependientes/genética , Anomalías Craneofaciales/genética , Anomalías del Ojo/genética , Trastornos del Crecimiento/genética , Luxación Congénita de la Cadera/genética , Proteínas Mitocondriales/genética , Osteocondrodisplasias/genética , Serina Proteasas/genética , Anomalías Dentarias/genética , Proteasas ATP-Dependientes/metabolismo , Adolescente , Animales , Línea Celular Tumoral , Niño , Preescolar , Variaciones en el Número de Copia de ADN , ADN Mitocondrial/genética , Exoma , Femenino , Frecuencia de los Genes , Células HEK293 , Células HeLa , Homocigoto , Humanos , Lactante , Masculino , Ratones , Microscopía Electrónica de Transmisión , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Mutación , Fenotipo , Estructura Terciaria de Proteína , Proteolisis , Serina Proteasas/metabolismo
9.
Biochim Biophys Acta Bioenerg ; 1858(7): 519-528, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28442264

RESUMEN

The mitochondrial ATP dependent matrix protease, Lon, is involved in the maintenance of mitochondrial DNA nucleoids and degradation of abnormal or misfolded proteins. The Lon protease regulates mitochondrial Tfam (mitochondrial transcription factor A) level and thus modulates mitochondrial DNA (mtDNA) content. We have previously shown that hypoxic stress induces the PKA-dependent phosphorylation of cytochrome c oxidase (CcO) subunits I, IVi1, and Vb and a time-dependent reduction of these subunits in RAW 264.7 murine macrophages subjected to hypoxia and rabbit hearts subjected to ischemia/reperfusion. Here, we show that Lon is involved in the preferential turnover of phosphorylated CcO subunits under hypoxic/ischemic stress. Induction of Lon protease occurs at 6 to 12 h of hypoxia and this increase coincides with lower CcO subunit contents. Over-expression of flag-tagged wild type and phosphorylation site mutant Vb and IVi1 subunits (S40A and T52A, respectively) caused marked degradation of wild type protein under hypoxia while the mutant proteins were relatively resistant. Furthermore, the recombinant purified Lon protease degraded the phosphorylated IVi1 and Vb subunits, while the phosphorylation-site mutant proteins were resistant to degradation. 3D structural modeling shows that the phosphorylation sites are exposed to the matrix compartment, accessible to matrix PKA and Lon protease. Hypoxic stress did not alter CcO subunit levels in Lon depleted cells, confirming its role in CcO turnover. Our results therefore suggest that Lon preferentially degrades the phosphorylated subunits of CcO and plays a role in the regulation of CcO activity in hypoxia and ischemia/reperfusion injury.


Asunto(s)
Proteasas ATP-Dependientes/metabolismo , Hipoxia de la Célula/fisiología , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias Cardíacas/enzimología , Proteínas Mitocondriales/metabolismo , Isquemia Miocárdica/enzimología , Proteasas ATP-Dependientes/química , Proteasas ATP-Dependientes/genética , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Masculino , Ratones , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Modelos Moleculares , Fosforilación , Conformación Proteica , Procesamiento Proteico-Postraduccional , Subunidades de Proteína , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/genética , Conejos , Proteínas Recombinantes/metabolismo
10.
Blood ; 119(14): 3321-9, 2012 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-22323447

RESUMEN

Synthetic triterpenoids are multitarget compounds exhibiting promise as preventative and therapeutic agents for cancer. Their proposed mechanism of action is by forming Michael adducts with reactive nucleophilic groups on target proteins. Our previous work demonstrates that the 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and its derivatives promote B-lymphoid cell apoptosis through a mitochondria-mediated pathway linked to mitochondrial protein aggregation. As one function of the Lon protease is to eliminate abnormal mitochondrial proteins, we hypothesized that CDDO-induced protein aggregation and lymphoma apoptosis occur by inactivating this enzyme. Here, we show that CDDO and its derivatives directly and selectively inhibit Lon. CDDO blocks Lon-mediated proteolysis in biochemical and cellular assays, but does not inhibit the 20S proteasome. Furthermore, a biotinylated-CDDO conjugate modifies mitochondrial Lon. A striking common phenotype of CDDO-treated lymphoma cells and Lon-knockdown cells is the accumulation of electron-dense aggregates within mitochondria. We also show that Lon protein levels are substantially elevated in malignant lymphoma cells, compared with resting or activated B cells. Finally, we demonstrate that Lon knockdown leads to lymphoma cell death. Together, these findings suggest that Lon inhibition plays a contributory role in CDDO-induced lymphoma cell death, and support the concept that mitochondrial Lon is a novel anticancer drug target.


Asunto(s)
Linfoma/enzimología , Mitocondrias/enzimología , Ácido Oleanólico/análogos & derivados , Proteasa La/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/ultraestructura , Linfoma/genética , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Ácido Oleanólico/síntesis química , Ácido Oleanólico/metabolismo , Ácido Oleanólico/farmacología , Proteasa La/antagonistas & inhibidores , Proteasa La/genética , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Unión Proteica , Regulación hacia Arriba
11.
Expert Opin Ther Targets ; 28(1-2): 9-15, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38156441

RESUMEN

INTRODUCTION: Mitochondrial LonP1 is an ATP-powered protease that also functions as an ATP-dependent chaperone. LonP1 plays a pivotal role in regulating mitochondrial proteostasis, metabolism and cell stress responses. Cancer cells exploit the functions of LonP1 to combat oncogenic stressors such as hypoxia, proteotoxicity, and oxidative stress, and to reprogram energy metabolism enabling cancer cell proliferation, chemoresistance, and metastasis. AREAS COVERED: LonP1 has emerged as a potential target for anti-cancer therapeutics. We review how cytoprotective functions of LonP1 can be leveraged by cancer cells to support oncogenic growth, proliferation, and survival. We also offer insights into small molecule inhibitors that target LonP1 by two distinct mechanisms: competitive inhibition of its protease activity and allosteric inhibition of its ATPase activity, both of which are crucial for its protease and chaperone functions. EXPERT OPINION: We highlight advantages of identifying specific, high-affinity allosteric inhibitors blocking the ATPase activity of LonP1. The future discovery of such inhibitors has potential application either alone or in conjunction with other anticancer agents, presenting an innovative approach and target for cancer therapeutics.


Asunto(s)
Metabolismo Energético , Péptido Hidrolasas , Humanos , Proliferación Celular , Adenosina Trifosfatasas , Adenosina Trifosfato
12.
Biochim Biophys Acta ; 1823(1): 56-66, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22119779

RESUMEN

The AAA(+) Lon protease is a soluble single-ringed homo-oligomer, which represents the most streamlined operational unit mediating ATP-dependent proteolysis. Despite its simplicity, the architecture of Lon proteases exhibits a species-specific diversity. Homology modeling provides insights into the structural features that distinguish bacterial and human Lon proteases as hexameric complexes from yeast Lon, which is uniquely heptameric. The best-understood functions of mitochondrial Lon are linked to maintaining proteostasis under normal metabolic conditions, and preventing proteotoxicity during environmental and cellular stress. An intriguing property of human Lon is its specific binding to G-quadruplex DNA, and its association with the mitochondrial genome in cultured cells. A fraction of Lon preferentially binds to the control region of mitochondrial DNA where transcription and replication are initiated. Here, we present an overview of the diverse functions of mitochondrial Lon, as well as speculative perspectives on its role in protein and mtDNA quality control.


Asunto(s)
Proteínas de Unión al ADN/química , Mitocondrias/enzimología , Proteasa La/química , Animales , Secuencia Conservada , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas de la Membrana , Datos de Secuencia Molecular , Proteasa La/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteolisis , Proteínas de Saccharomyces cerevisiae
13.
Neurology ; 101(15): e1567-e1571, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37460232

RESUMEN

Pathogenic biallelic variants in ACO2, which encodes the enzyme mitochondrial aconitase, are associated with the very rare diagnosis of ACO2-related infantile cerebellar retinal degeneration (OMIM 614559). We describe the diagnostic odyssey of a 4-year-old female patient with profound global developmental delays, microcephaly, severe hypotonia, retinal dystrophy, seizures, and progressive cerebellar atrophy. Whole-exome sequencing revealed 2 variants in ACO2; c.2105_2106delAG (p.Gln702ArgfsX9), a likely pathogenic variant, and c.988C>T (p.Pro330Ser) which was classified as a variant of uncertain significance (VUS). While the VUS was confirmed to be maternally inherited, the phase of the other variant could not be confirmed due to lack of a paternal sample. Functional biochemical studies were performed on a research basis to clarify the interpretation of the VUS, which enabled clinical confirmation of the diagnosis of ACO2-related infantile cerebellar retinal degeneration for our patient.


Asunto(s)
Microcefalia , Malformaciones del Sistema Nervioso , Distrofias Retinianas , Femenino , Humanos , Niño , Preescolar , Aconitato Hidratasa , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/genética , Atrofia
14.
Research (Wash D C) ; 6: 0175, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37333972

RESUMEN

Interorganelle contacts and communications are increasingly recognized to play a vital role in cellular function and homeostasis. In particular, the mitochondria-endoplasmic reticulum (ER) membrane contact site (MAM) is known to regulate ion and lipid transfer, as well as signaling and organelle dynamics. However, the regulatory mechanisms of MAM formation and their function are still elusive. Here, we identify mitochondrial Lon protease (LonP1), a highly conserved mitochondrial matrix protease, as a new MAM tethering protein. The removal of LonP1 substantially reduces MAM formation and causes mitochondrial fragmentation. Furthermore, deletion of LonP1 in the cardiomyocytes of mouse heart impairs MAM integrity and mitochondrial fusion and activates the unfolded protein response within the ER (UPRER). Consequently, cardiac-specific LonP1 deficiency causes aberrant metabolic reprogramming and pathological heart remodeling. These findings demonstrate that LonP1 is a novel MAM-localized protein orchestrating MAM integrity, mitochondrial dynamics, and UPRER, offering exciting new insights into the potential therapeutic strategy for heart failure.

15.
Nucleic Acids Res ; 38(19): 6466-76, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20530535

RESUMEN

Mammalian mitochondria contain full-length genome and a single-stranded 7S DNA. Although the copy number of mitochondrial DNA (mtDNA) varies depending on the cell type and also in response to diverse environmental stresses, our understanding of how mtDNA and 7S DNA are maintained and regulated is limited, partly due to lack of reliable in vitro assay systems that reflect the in vivo functionality of mitochondria. Here we report an in vitro assay system to measure synthesis of both mtDNA and 7S DNA under a controllable in vitro condition. With this assay system, we demonstrate that the replication capacity of mitochondria correlates with endogenous copy numbers of mtDNA and 7S DNA. Our study also shows that higher nucleotide concentrations increasingly promote 7S DNA synthesis but not mtDNA synthesis. Consistently, the mitochondrial capacity to synthesize 7S DNA but not mtDNA noticeably varied along the cell cycle, reaching its highest level in S phase. These findings suggest that syntheses of mtDNA and 7S DNA proceed independently and that the mitochondrial capacity to synthesize 7S DNA dynamically changes not only with cell-cycle progression but also in response to varying nucleotide concentrations.


Asunto(s)
Ciclo Celular/genética , ADN Mitocondrial/biosíntesis , ADN de Cadena Simple/biosíntesis , Genoma Mitocondrial , Variaciones en el Número de Copia de ADN , ADN Polimerasa gamma , ADN Polimerasa Dirigida por ADN/metabolismo , Genoma Humano , Células HeLa , Humanos , Proteínas Mitocondriales/análisis , Nucleótidos/metabolismo
16.
Nat Metab ; 4(6): 739-758, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35760869

RESUMEN

Mitochondria are the main consumers of oxygen within the cell. How mitochondria sense oxygen levels remains unknown. Here we show an oxygen-sensitive regulation of TFAM, an activator of mitochondrial transcription and replication, whose alteration is linked to tumours arising in the von Hippel-Lindau syndrome. TFAM is hydroxylated by EGLN3 and subsequently bound by the von Hippel-Lindau tumour-suppressor protein, which stabilizes TFAM by preventing mitochondrial proteolysis. Cells lacking wild-type VHL or in which EGLN3 is inactivated have reduced mitochondrial mass. Tumorigenic VHL variants leading to different clinical manifestations fail to bind hydroxylated TFAM. In contrast, cells harbouring the Chuvash polycythaemia VHLR200W mutation, involved in hypoxia-sensing disorders without tumour development, are capable of binding hydroxylated TFAM. Accordingly, VHL-related tumours, such as pheochromocytoma and renal cell carcinoma cells, display low mitochondrial content, suggesting that impaired mitochondrial biogenesis is linked to VHL tumorigenesis. Finally, inhibiting proteolysis by targeting LONP1 increases mitochondrial content in VHL-deficient cells and sensitizes therapy-resistant tumours to sorafenib treatment. Our results offer pharmacological avenues to sensitize therapy-resistant VHL tumours by focusing on the mitochondria.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Enfermedad de von Hippel-Lindau , Proteasas ATP-Dependientes , Carcinoma de Células Renales/genética , Humanos , Neoplasias Renales/genética , Proteínas Mitocondriales , Biogénesis de Organelos , Oxígeno , Enfermedad de von Hippel-Lindau/genética
17.
Nucleic Acids Res ; 36(4): 1273-87, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18174225

RESUMEN

Lon is an ATP-powered protease that binds DNA. However, the function of DNA binding by Lon remains elusive. Studies suggest that human Lon (hLon) binds preferentially to a G-rich single-stranded DNA (ssDNA) sequence overlapping the light strand promoter of mitochondrial DNA. This sequence is contained within a 24-base oligonucleotide referred to as LSPas. Here, we use biochemical and biophysical approaches to elucidate the structural properties of ssDNAs bound by hLon, as well as the thermodynamics of DNA binding by hLon. Electrophoretic mobility shift assay and circular dichroism show that ssDNAs with a propensity for forming parallel G-quartets are specifically bound by hLon. Isothermal titration calorimetry demonstrates that hLon binding to LSPas is primarily driven by enthalpy change associated with a significant reduction in heat capacity. Differential scanning calorimetry pinpoints an excess heat capacity upon hLon binding to LSPas. By contrast, hLon binding to an 8-base G-rich core sequence is entropically driven with a relatively negligible change in heat capacity. A considerable enhancement of thermal stability accompanies hLon binding to LSPas as compared to the G-rich core. Taken together, these data support the notion that hLon binds G-quartets through rigid-body binding and that binding to LSPas is coupled with structural adaptation.


Asunto(s)
ADN Mitocondrial/química , ADN de Cadena Simple/química , Proteínas de Unión al ADN/química , G-Cuádruplex , Proteasa La/química , Termodinámica , Sitios de Unión , Calorimetría , ADN Mitocondrial/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Calor , Humanos , Proteasa La/metabolismo , Unión Proteica
18.
Mitochondrion ; 51: 46-61, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31756517

RESUMEN

Mitochondrial LonP1 is an essential stress response protease that mediates mitochondrial proteostasis, metabolism and bioenergetics. Homozygous and compound heterozygous variants in the LONP1 gene encoding the LonP1 protease have recently been shown to cause a diverse spectrum of human pathologies, ranging from classical mitochondrial disease phenotypes, profound neurologic impairment and multi-organ dysfunctions, some of which are uncommon to mitochondrial disorders. In this review, we focus primarily on human LonP1 and discuss findings, which demonstrate its multidimensional roles in maintaining mitochondrial proteostasis and adapting cells to metabolic flux and stress during normal physiology and disease processes. We also discuss emerging roles of LonP1 in responding to developmental, oncogenic and cardiac stress.


Asunto(s)
Proteasas ATP-Dependientes/metabolismo , Metabolismo Energético/fisiología , Mitocondrias/metabolismo , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/metabolismo , Estrés Fisiológico/fisiología , Proteasas ATP-Dependientes/genética , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/fisiología , Estrés del Retículo Endoplásmico/fisiología , Humanos , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Mutación Missense/genética , Respuesta de Proteína Desplegada/fisiología
19.
MedEdPORTAL ; 16: 10934, 2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32704540

RESUMEN

Introduction: As evidenced by student performance on various assessments, pharmacotherapy remains a comparative weakness in undergraduate medical education, with several institutions developing novel strategies for students to apply these principles in a practical setting. Medical curricula have recently prioritized group-learning modalities and evidence-based medicine education. However, these principles have yet to impact pharmacology education. We developed and implemented an evidence-based, group-learning exercise for first-year medical students focusing on pharmacology through the practical lens of pharmacotherapy and pharmacopolicy. Methods: First-year medical students in different groups were assigned a particular medication and, during an in-class session, were encouraged to meet with other representatives assigned the same drug to interpret the provided package insert and any online information. Students then reconvened with their groups to engage in collaborative teaching about each assigned drug before completing a group quiz using online resources. Facilitators reviewed the group quiz and allowed time for student questions. Results: For 180 participants, the average group-quiz score was 86%, ranging from 68% to 100%. Student-reported satisfaction with the activity in meeting its preset objectives averaged 3.7 on a 5-point scale, with 5 being most positive. Discussion: Overall, this activity effectively integrates principles of pharmacotherapy and pharmacopolicy into a group-based, evidence-based exercise. Limitations of the activity include the number of possible example drugs and the amount of material covered in a given time frame. However, the activity lends itself to the role of an introductory session in a longer curriculum centered on clinical-applied pharmacology and evidence-based practice.


Asunto(s)
Educación de Pregrado en Medicina , Estudiantes de Medicina , Curriculum , Medicina Basada en la Evidencia , Humanos , Aprendizaje
20.
Biochim Biophys Acta ; 1784(5): 727-35, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18359303

RESUMEN

Lon, also known as the protease La, is a homo-oligomeric ATP-dependent protease, which is highly conserved in archaea, eubacteria and eukaryotic mitochondria and peroxisomes. Since its discovery, studies have shown that Lon activity is essential for cellular homeostasis, mediating protein quality control and metabolic regulation. This article highlights the discoveries made over the past decade demonstrating that Lon selectively degrades abnormal as well as certain regulatory proteins and thus plays significant roles in maintaining bacterial and mitochondrial function and integrity. In addition, Lon is required in certain pathogenic bacteria, for rendering pathogenicity and host infectivity. Recent research endeavors have been directed toward elucidating the reaction mechanism of the Lon protease by different biochemical and structural biological techniques. In this mini-review, the authors survey the diverse biological roles of Lon, and also place special emphasis on recent findings that clarify the mechanistic aspects of the Lon reaction cycle.


Asunto(s)
Péptidos/metabolismo , Proteasa La/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Proteínas de Unión al ADN/metabolismo , Datos de Secuencia Molecular , Péptidos/química , Proteasa La/química , Procesamiento Proteico-Postraduccional , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA