Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 20(7): 812-822, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38167917

RESUMEN

Sirtuins are nicotinamide adenine dinucleotide (NAD+)-dependent protein lysine deacylases regulating metabolism and stress responses; however, characterization of the removed acyl groups and their downstream metabolic fates remains incomplete. Here we employed untargeted comparative metabolomics to reinvestigate mitochondrial sirtuin biochemistry. First, we identified N-glutarylspermidines as metabolites downstream of the mitochondrial sirtuin SIR-2.3 in Caenorhabditis elegans and demonstrated that SIR-2.3 functions as a lysine deglutarylase and that N-glutarylspermidines can be derived from O-glutaryl-ADP-ribose. Subsequent targeted analysis of C. elegans, mouse and human metabolomes revealed a chemically diverse range of N-acylspermidines, and formation of N-succinylspermidines and/or N-glutarylspermidines was observed downstream of mammalian mitochondrial sirtuin SIRT5 in two cell lines, consistent with annotated functions of SIRT5. Finally, N-glutarylspermidines were found to adversely affect C. elegans lifespan and mammalian cell proliferation. Our results indicate that N-acylspermidines are conserved metabolites downstream of mitochondrial sirtuins that facilitate annotation of sirtuin enzymatic activities in vivo and may contribute to sirtuin-dependent phenotypes.


Asunto(s)
Caenorhabditis elegans , Mitocondrias , Sirtuinas , Sirtuinas/metabolismo , Caenorhabditis elegans/metabolismo , Animales , Mitocondrias/metabolismo , Humanos , Ratones , Proliferación Celular , Metabolómica
2.
Proc Natl Acad Sci U S A ; 119(11): e2117013119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35259022

RESUMEN

SignificanceThe study provided a long-sought molecular mechanism that could explain the link between fatty acid metabolism and cancer metastasis. Further understanding may lead to new strategies to inhibit cancer metastasis. The chemical proteomic approach developed here will be useful for discovering other regulatory mechanisms of protein function by small molecule metabolites.


Asunto(s)
Acilcoenzima A/metabolismo , Nucleósido Difosfato Quinasas NM23/antagonistas & inhibidores , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias de la Mama , Endocitosis , Femenino , Humanos , Metástasis de la Neoplasia , Neoplasias/etiología , Unión Proteica , Proteoma , Proteómica/métodos
3.
Proc Natl Acad Sci U S A ; 119(45): e2200477119, 2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36322753

RESUMEN

IGF2BP2 binds to a number of RNA transcripts and has been suggested to function as a tumor promoter, although little is known regarding the mechanisms that regulate its roles in RNA metabolism. Here we demonstrate that IGF2BP2 binds to the 3' untranslated region of the transcript encoding ATP6V1A, a catalytic subunit of the vacuolar ATPase (v-ATPase), and serves as a substrate for the NAD+-dependent deacetylase SIRT1, which regulates how IGF2BP2 affects the stability of the ATP6V1A transcript. When sufficient levels of SIRT1 are expressed, it catalyzes the deacetylation of IGF2BP2, which can bind to the ATP6V1A transcript but does not mediate its degradation. However, when SIRT1 expression is low, the acetylated form of IGF2BP2 accumulates, and upon binding to the ATP6V1A transcript recruits the XRN2 nuclease, which catalyzes transcript degradation. Thus, the stability of the ATP6V1A transcript is significantly compromised in breast cancer cells when SIRT1 expression is low or knocked-down. This leads to a reduction in the expression of functional v-ATPase complexes in cancer cells and to an impairment in their lysosomal activity, resulting in the production of a cellular secretome consisting of increased numbers of exosomes enriched in ubiquitinated protein cargo and soluble hydrolases, including cathepsins, that together combine to promote tumor cell survival and invasiveness. These findings describe a previously unrecognized role for IGF2BP2 in mediating the degradation of a messenger RNA transcript essential for lysosomal function and highlight how its sirtuin-regulated acetylation state can have significant biological and disease consequences.


Asunto(s)
Neoplasias , ATPasas de Translocación de Protón Vacuolares , Humanos , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Sirtuina 1/metabolismo , ARN/metabolismo , Procesos Neoplásicos , Lisosomas/genética , Lisosomas/metabolismo , Neoplasias/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
4.
Br J Cancer ; 127(9): 1577-1583, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36229581

RESUMEN

Germ cell tumours (GCTs) are a heterogeneous group of rare neoplasms that present in different anatomical sites and across a wide spectrum of patient ages from birth through to adulthood. Once these strata are applied, cohort numbers become modest, hindering inferences regarding management and therapeutic advances. Moreover, patients with GCTs are treated by different medical professionals including paediatric oncologists, neuro-oncologists, medical oncologists, neurosurgeons, gynaecological oncologists, surgeons, and urologists. Silos of care have thus formed, further hampering knowledge dissemination between specialists. Dedicated biobank specimen collection is therefore critical to foster continuous growth in our understanding of similarities and differences by age, gender, and site, particularly for rare cancers such as GCTs. Here, the Malignant Germ Cell International Consortium provides a framework to create a sustainable, global research infrastructure that facilitates acquisition of tissue and liquid biopsies together with matched clinical data sets that reflect the diversity of GCTs. Such an effort would create an invaluable repository of clinical and biological data which can underpin international collaborations that span professional boundaries, translate into clinical practice, and ultimately impact patient outcomes.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias , Neoplasias Testiculares , Niño , Humanos , Adulto , Masculino , Investigación Biomédica Traslacional , Neoplasias de Células Germinales y Embrionarias/terapia , Neoplasias Testiculares/patología
5.
Proc Natl Acad Sci U S A ; 116(52): 26625-26632, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31843902

RESUMEN

The mitochondrial enzyme glutaminase (GLS) is frequently up-regulated during tumorigenesis and is being evaluated as a target for cancer therapy. GLS catalyzes the hydrolysis of glutamine to glutamate, which then supplies diverse metabolic pathways with carbon and/or nitrogen. Here, we report that SIRT5, a mitochondrial NAD+-dependent lysine deacylase, plays a key role in stabilizing GLS. In transformed cells, SIRT5 regulates glutamine metabolism by desuccinylating GLS and thereby protecting it from ubiquitin-mediated degradation. Moreover, we show that SIRT5 is up-regulated during cellular transformation and supports proliferation and tumorigenesis. Elevated SIRT5 expression in human breast tumors correlates with poor patient prognosis. These findings reveal a mechanism for increasing GLS expression in cancer cells and establish a role for SIRT5 in metabolic reprogramming and mammary tumorigenesis.

6.
FASEB J ; 34(1): 1846-1858, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914635

RESUMEN

P53 has been implicated in the pathogenesis of obesity and diabetes; however, the mechanisms and tissue sites of action are incompletely defined. Therefore, we investigated the role of hepatocyte p53 in metabolic homeostasis using a hepatocyte-specific p53 knockout mouse model. To gain further mechanistic insight, we studied mice under two complementary conditions of restricted weight gain: vertical sleeve gastrectomy (VSG) or food restriction. VSG or sham surgery was performed in high-fat diet-fed male hepatocyte-specific p53 wild-type and knockout littermates. Sham-operated mice were fed ad libitum or food restricted to match their body weight to VSG-operated mice. Hepatocyte-specific p53 ablation in sham-operated ad libitum-fed mice impaired glucose homeostasis, increased body weight, and decreased energy expenditure without changing food intake. The metabolic deficits induced by hepatocyte-specific p53 ablation were corrected, in part by food restriction, and completely by VSG. Unlike food restriction, VSG corrected the effect of hepatocyte p53 ablation to lower energy expenditure, resulting in a greater improvement in glucose homeostasis compared with food restricted mice. These data reveal an important new role for hepatocyte p53 in the regulation of energy expenditure and body weight and suggest that VSG can improve alterations in energetics associated with p53 dysregulation.


Asunto(s)
Hepatocitos/metabolismo , Enfermedades Metabólicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/fisiología , Restricción Calórica/métodos , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Alimentos , Gastrectomía/métodos , Homeostasis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Aumento de Peso/fisiología , Pérdida de Peso
7.
J Biol Chem ; 294(43): 15623-15637, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31434739

RESUMEN

Defects in the Fanconi anemia (FA) DNA damage-response pathway result in genomic instability, developmental defects, hematopoietic failure, cancer predisposition, and metabolic disorders. The endogenous sources of damage contributing to FA phenotypes and the links between FA and metabolic disease remain poorly understood. Here, using mice lacking the Fancd2 gene, encoding a central FA pathway component, we investigated whether the FA pathway protects against metabolic challenges. Fancd2-/- and wildtype (WT) mice were fed a standard diet (SD), a diet enriched in fat, cholesterol, and cholic acid (Paigen diet), or a diet enriched in lipid alone (high-fat diet (HFD)). Fancd2-/- mice developed hepatobiliary disease and exhibited decreased survival when fed a Paigen diet but not a HFD. Male Paigen diet-fed mice lacking Fancd2 had significant biliary hyperplasia, increased serum bile acid concentration, and increased hepatic pathology. In contrast, female mice were similarly impacted by Paigen diet feeding regardless of Fancd2 status. Upon Paigen diet challenge, male Fancd2-/- mice had altered expression of genes encoding hepatic bile acid transporters and cholesterol and fatty acid metabolism proteins, including Scp2/x, Abcg5/8, Abca1, Ldlr, Srebf1, and Scd-1 Untargeted lipidomic profiling in liver tissue revealed 132 lipid species, including sphingolipids, glycerophospholipids, and glycerolipids, that differed significantly in abundance depending on Fancd2 status in male mice. We conclude that the FA pathway has sex-specific impacts on hepatic lipid and bile acid metabolism, findings that expand the known functions of the FA pathway and may provide mechanistic insight into the metabolic disease predisposition in individuals with FA.


Asunto(s)
Bilis/metabolismo , Dieta , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Metabolismo de los Lípidos , Hígado/metabolismo , Caracteres Sexuales , Animales , Colesterol/metabolismo , Daño del ADN , Enfermedades del Sistema Digestivo/metabolismo , Susceptibilidad a Enfermedades , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Conducta Alimentaria , Femenino , Regulación de la Expresión Génica , Cinética , Metabolismo de los Lípidos/genética , Masculino , Ratones
8.
Nat Chem Biol ; 14(10): 943-954, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30150681

RESUMEN

Since the origins of DNA-based life, the enzyme ribonucleotide reductase (RNR) has spurred proliferation because of its rate-limiting role in de novo deoxynucleoside-triphosphate (dNTP) biosynthesis. Paradoxically, the large subunit, RNR-α, of this obligatory two-component complex in mammals plays a context-specific antiproliferative role. There is little explanation for this dichotomy. Here, we show that RNR-α has a previously unrecognized DNA-replication inhibition function, leading to growth retardation. This underappreciated biological activity functions in the nucleus, where RNR-α interacts with ZRANB3. This process suppresses ZRANB3's function in unstressed cells, which we show to promote DNA synthesis. This nonreductase function of RNR-α is promoted by RNR-α hexamerization-induced by a natural and synthetic nucleotide of dA/ClF/CLA/FLU-which elicits rapid RNR-α nuclear import. The newly discovered nuclear signaling axis is a primary defense against elevated or imbalanced dNTP pools that can exert mutagenic effects irrespective of the cell cycle.


Asunto(s)
Núcleo Celular/metabolismo , ADN Helicasas/antagonistas & inhibidores , Mutación , Ribonucleótido Reductasas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Células COS , Ciclo Celular , Proliferación Celular , Chlorocebus aethiops , Citosol/metabolismo , ADN/análisis , Daño del ADN , Replicación del ADN , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Células K562 , Ratones , Mutagénesis , Células 3T3 NIH , Unión Proteica , ARN Interferente Pequeño/metabolismo , Transducción de Señal
9.
Proc Natl Acad Sci U S A ; 114(3): 492-497, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28034921

RESUMEN

Tumor microvasculature tends to be malformed, more permeable, and more tortuous than vessels in healthy tissue, effects that have been largely attributed to up-regulated VEGF expression. However, tumor tissue tends to stiffen during solid tumor progression, and tissue stiffness is known to alter cell behaviors including proliferation, migration, and cell-cell adhesion, which are all requisite for angiogenesis. Using in vitro, in vivo, and ex ovo models, we investigated the effects of matrix stiffness on vessel growth and integrity during angiogenesis. Our data indicate that angiogenic outgrowth, invasion, and neovessel branching increase with matrix cross-linking. These effects are caused by increased matrix stiffness independent of matrix density, because increased matrix density results in decreased angiogenesis. Notably, matrix stiffness up-regulates matrix metalloproteinase (MMP) activity, and inhibiting MMPs significantly reduces angiogenic outgrowth in stiffer cross-linked gels. To investigate the functional significance of altered endothelial cell behavior in response to matrix stiffness, we measured endothelial cell barrier function on substrates mimicking the stiffness of healthy and tumor tissue. Our data indicate that barrier function is impaired and the localization of vascular endothelial cadherin is altered as function of matrix stiffness. These results demonstrate that matrix stiffness, separately from matrix density, can alter vascular growth and integrity, mimicking the changes that exist in tumor vasculature. These data suggest that therapeutically targeting tumor stiffness or the endothelial cell response to tumor stiffening may help restore vessel structure, minimize metastasis, and aid in drug delivery.


Asunto(s)
Matriz Extracelular/fisiología , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/fisiopatología , Microvasos/fisiopatología , Animales , Fenómenos Biomecánicos , Bovinos , Células Cultivadas , Embrión de Pollo , Colágeno/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Mamarias Experimentales/patología , Metaloproteinasas de la Matriz/metabolismo , Ratones , Microvasos/patología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Fenotipo , Microambiente Tumoral/fisiología , Rigidez Vascular/fisiología
10.
Proc Natl Acad Sci U S A ; 113(16): 4320-5, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27051063

RESUMEN

Cellular metabolites, such as acyl-CoA, can modify proteins, leading to protein posttranslational modifications (PTMs). One such PTM is lysine succinylation, which is regulated by sirtuin 5 (SIRT5). Although numerous proteins are modified by lysine succinylation, the physiological significance of lysine succinylation and SIRT5 remains elusive. Here, by profiling acyl-CoA molecules in various mouse tissues, we have discovered that different tissues have different acyl-CoA profiles and that succinyl-CoA is the most abundant acyl-CoA molecule in the heart. This interesting observation has prompted us to examine protein lysine succinylation in different mouse tissues in the presence and absence of SIRT5. Protein lysine succinylation predominantly accumulates in the heart whenSirt5is deleted. Using proteomic studies, we have identified many cardiac proteins regulated by SIRT5. Our data suggest that ECHA, a protein involved in fatty acid oxidation, is a major enzyme that is regulated by SIRT5 and affects heart function.Sirt5knockout (KO) mice have lower ECHA activity, increased long-chain acyl-CoAs, and decreased ATP in the heart under fasting conditions.Sirt5KO mice develop hypertrophic cardiomyopathy, as evident from the increased heart weight relative to body weight, as well as reduced shortening and ejection fractions. These findings establish that regulating heart metabolism and function is a major physiological function of lysine succinylation and SIRT5.


Asunto(s)
Acilcoenzima A/metabolismo , Cardiomegalia/metabolismo , Ácidos Grasos/metabolismo , Miocardio/metabolismo , Procesamiento Proteico-Postraduccional , Sirtuinas/metabolismo , Acilcoenzima A/genética , Acilación , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Ácidos Grasos/genética , Metabolómica/métodos , Ratones , Ratones Noqueados , Miocardio/patología , Oxidación-Reducción , Proteómica/métodos , Sirtuinas/genética
11.
Biochem Soc Trans ; 46(6): 1643-1651, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30514768

RESUMEN

Eukaryotic cells continuously experience DNA damage that can perturb key molecular processes like DNA replication. DNA replication forks that encounter DNA lesions typically slow and may stall, which can lead to highly detrimental fork collapse if appropriate protective measures are not executed. Stabilization and protection of stalled replication forks ensures the possibility of effective fork restart and prevents genomic instability. Recent efforts from multiple laboratories have highlighted several proteins involved in replication fork remodeling and DNA damage response pathways as key regulators of fork stability. Homologous recombination factors such as RAD51, BRCA1, and BRCA2, along with components of the Fanconi Anemia pathway, are now known to be crucial for stabilizing stalled replication forks and preventing nascent strand degradation. Several checkpoint proteins have additionally been implicated in fork protection. Ongoing work in this area continues to shed light on a sophisticated molecular pathway that balances the action of DNA resection and fork protection to maintain genomic integrity, with important implications for the fate of both normal and malignant cells following replication stress.


Asunto(s)
Daño del ADN/genética , Animales , Replicación del ADN/genética , Replicación del ADN/fisiología , Inestabilidad Genómica/genética , Inestabilidad Genómica/fisiología , Humanos
12.
Proc Natl Acad Sci U S A ; 112(27): 8314-9, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26106154

RESUMEN

Alternative splicing of proteins gives rise to different isoforms that play a crucial role in regulating several cellular processes. Notably, splicing profiles are altered in several cancer types, and these profiles are believed to be involved in driving the oncogenic process. Although the importance of alternative splicing alterations occurring during cancer is increasingly appreciated, the underlying regulatory mechanisms remain poorly understood. In this study, we use both biochemical and physical tools coupled with engineered models, patient samples, and a murine model to investigate the role of the mechanical properties of the tumor microenvironment in regulating the production of the extra domain-B (EDB) splice variant of fibronectin (FN), a hallmark of tumor angiogenesis. Specifically, we show that the amount of EDB-FN produced by endothelial cells increases with matrix stiffness both in vitro and within mouse mammary tumors. Matrix stiffness regulates splicing through the activation of serine/arginine rich (SR) proteins, the splicing factors involved in the production of FN isoforms. Activation of the SR proteins by matrix stiffness and the subsequent production of EDB-FN are dependent on intracellular contractility and PI3K-AKT signaling. Notably, matrix stiffness-mediated splicing is not limited to EDB-FN, but also affects splicing in the production of PKC ßII and the VEGF 165b splice variant. Together, these results demonstrate that the mechanical properties of the microenvironment regulate alternative splicing and establish a previously unidentified mechanism by which cells can adapt to their microenvironment.


Asunto(s)
Empalme Alternativo , Fibronectinas/genética , Neoplasias/genética , Microambiente Tumoral/genética , Animales , Arginina/genética , Arginina/metabolismo , Sitios de Unión/genética , Fenómenos Biomecánicos , Western Blotting , Bovinos , Células Cultivadas , Células Endoteliales/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Humanos , Ratones , Microscopía Confocal , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Serina/genética , Serina/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
13.
J Biol Chem ; 291(31): 16011-23, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27467582

RESUMEN

Li-Fraumeni syndrome (LFS) patients harbor germ line mutations in the TP53 gene and are at increased risk of hormone receptor-positive breast cancers. Recently, elevated levels of aromatase, the rate-limiting enzyme for estrogen biosynthesis, were found in the breast tissue of LFS patients. Although p53 down-regulates aromatase expression, the underlying mechanisms are incompletely understood. In the present study, we found that LFS stromal cells expressed higher levels of Hsp90 ATPase activity and aromatase compared with wild-type stromal cells. Inhibition of Hsp90 ATPase suppressed aromatase expression. Silencing Aha1 (activator of Hsp90 ATPase 1), a co-chaperone of Hsp90 required for its ATPase activity, led to both inhibition of Hsp90 ATPase activity and reduced aromatase expression. In comparison with wild-type stromal cells, increased levels of the Hsp90 client proteins, HIF-1α, and PKM2 were found in LFS stromal cells. A complex comprised of HIF-1α and PKM2 was recruited to the aromatase promoter II in LFS stromal cells. Silencing either HIF-1α or PKM2 suppressed aromatase expression in LFS stromal cells. CP-31398, a p53 rescue compound, suppressed levels of Aha1, Hsp90 ATPase activity, levels of PKM2 and HIF-1α, and aromatase expression in LFS stromal cells. Consistent with these in vitro findings, levels of Hsp90 ATPase activity, Aha1, HIF-1α, PKM2, and aromatase were increased in the mammary glands of p53 null versus wild-type mice. PKM2 and HIF-1α were shown to co-localize in the nucleus of stromal cells of LFS breast tissue. Taken together, our results show that the Aha1-Hsp90-PKM2/HIF-1α axis mediates the induction of aromatase in LFS.


Asunto(s)
Tejido Adiposo/metabolismo , Aromatasa/biosíntesis , Mama/metabolismo , Proteínas Portadoras/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Síndrome de Li-Fraumeni/metabolismo , Glándulas Mamarias Animales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Hormonas Tiroideas/metabolismo , Tejido Adiposo/patología , Animales , Aromatasa/genética , Mama/patología , Proteínas Portadoras/genética , Línea Celular , Femenino , Proteínas HSP90 de Choque Térmico/genética , Humanos , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/patología , Glándulas Mamarias Animales/patología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas de Neoplasias/genética , Células del Estroma/metabolismo , Células del Estroma/patología , Hormonas Tiroideas/genética , Proteínas de Unión a Hormona Tiroide
17.
J Biol Chem ; 290(24): 14826-40, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25911100

RESUMEN

The RAD9A-HUS1-RAD1 (9-1-1) complex is a heterotrimeric clamp that promotes checkpoint signaling and repair at DNA damage sites. In this study, we elucidated HUS1 functional residues that drive clamp assembly, DNA interactions, and downstream effector functions. First, we mapped a HUS1-RAD9A interface residue that was critical for 9-1-1 assembly and DNA loading. Next, we identified multiple positively charged residues in the inner ring of HUS1 that were crucial for genotoxin-induced 9-1-1 chromatin localization and ATR signaling. Finally, we found two hydrophobic pockets on the HUS1 outer surface that were important for cell survival after DNA damage. Interestingly, these pockets were not required for 9-1-1 chromatin localization or ATR-mediated CHK1 activation but were necessary for interactions between HUS1 and its binding partner MYH, suggesting that they serve as interaction domains for the recruitment and coordination of downstream effectors at damage sites. Together, these results indicate that, once properly loaded onto damaged DNA, the 9-1-1 complex executes multiple, separable functions that promote genome maintenance.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN/metabolismo , Genoma Humano , Transducción de Señal , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Secuencia de Bases , Proteínas de Ciclo Celular/química , Células Cultivadas , Cartilla de ADN , Humanos , Ratones , Conformación Proteica
18.
PLoS Genet ; 9(2): e1003320, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23468651

RESUMEN

The RAD9-RAD1-HUS1 (9-1-1) complex is a heterotrimeric PCNA-like clamp that responds to DNA damage in somatic cells by promoting DNA repair as well as ATR-dependent DNA damage checkpoint signaling. In yeast, worms, and flies, the 9-1-1 complex is also required for meiotic checkpoint function and efficient completion of meiotic recombination; however, since Rad9, Rad1, and Hus1 are essential genes in mammals, little is known about their functions in mammalian germ cells. In this study, we assessed the meiotic functions of 9-1-1 by analyzing mice with germ cell-specific deletion of Hus1 as well as by examining the localization of RAD9 and RAD1 on meiotic chromosomes during prophase I. Hus1 loss in testicular germ cells resulted in meiotic defects, germ cell depletion, and severely compromised fertility. Hus1-deficient primary spermatocytes exhibited persistent autosomal γH2AX and RAD51 staining indicative of unrepaired meiotic DSBs, synapsis defects, an extended XY body domain often encompassing partial or whole autosomes, and an increase in structural chromosome abnormalities such as end-to-end X chromosome-autosome fusions and ruptures in the synaptonemal complex. Most of these aberrations persisted in diplotene-stage spermatocytes. Consistent with a role for the 9-1-1 complex in meiotic DSB repair, RAD9 localized to punctate, RAD51-containing foci on meiotic chromosomes in a Hus1-dependent manner. Interestingly, RAD1 had a broader distribution that only partially overlapped with RAD9, and localization of both RAD1 and the ATR activator TOPBP1 to the XY body and to unsynapsed autosomes was intact in Hus1 conditional knockouts. We conclude that mammalian HUS1 acts as a component of the canonical 9-1-1 complex during meiotic prophase I to promote DSB repair and further propose that RAD1 and TOPBP1 respond to unsynapsed chromatin through an alternative mechanism that does not require RAD9 or HUS1.


Asunto(s)
Proteínas de Ciclo Celular , Cromosomas/genética , Exonucleasas , Meiosis/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Daño del ADN/genética , Daño del ADN/fisiología , Reparación del ADN/genética , Exonucleasas/genética , Exonucleasas/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Ratones , Complejos Multiproteicos , Testículo/citología , Testículo/metabolismo
19.
Proc Natl Acad Sci U S A ; 110(15): 6187-92, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23530224

RESUMEN

We previously demonstrated that cardiac myosin can use 2-deoxy-ATP (dATP) as an energy substrate, that it enhances contraction and relaxation with minimal effect on calcium-handling properties in vitro, and that contractile enhancement occurs with only minor elevation of cellular [dATP]. Here, we report the effect of chronically enhanced dATP concentration on cardiac function using a transgenic mouse that overexpresses the enzyme ribonucleotide reductase (TgRR), which catalyzes the rate-limiting step in de novo deoxyribonucleotide biosynthesis. Hearts from TgRR mice had elevated left ventricular systolic function compared with wild-type (WT) mice, both in vivo and in vitro, without signs of hypertrophy or altered diastolic function. Isolated cardiomyocytes from TgRR mice had enhanced contraction and relaxation, with no change in Ca(2+) transients, suggesting targeted improvement of myofilament function. TgRR hearts had normal ATP and only slightly decreased phosphocreatine levels by (31)P NMR spectroscopy, and they maintained rate responsiveness to dobutamine challenge. These data demonstrate long-term (at least 5-mo) elevation of cardiac [dATP] results in sustained elevation of basal left ventricular performance, with maintained ß-adrenergic responsiveness and energetic reserves. Combined with results from previous studies, we conclude that this occurs primarily via enhanced myofilament activation and contraction, with similar or faster ability to relax. The data are sufficiently compelling to consider elevated cardiac [dATP] as a therapeutic option to treat systolic dysfunction.


Asunto(s)
Regulación de la Expresión Génica , Miocardio/metabolismo , Ribonucleótido Reductasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Ecocardiografía , Espectroscopía de Resonancia Magnética , Ratones , Ratones Transgénicos , Miocitos Cardíacos/citología , Fenotipo , Ribonucleótido Reductasas/genética , Sarcómeros/metabolismo , Sístole , Transgenes
20.
J Biol Chem ; 289(10): 6513-6525, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24451373

RESUMEN

The p53 tumor suppressor gene encodes a homotetrameric transcription factor which is activated in response to a variety of cellular stressors, including DNA damage and oncogene activation. p53 mutations occur in >50% of human cancers. Although p53 has been shown to regulate Wnt signaling, the underlying mechanisms are not well understood. Here we show that silencing p53 in colon cancer cells led to increased expression of Aha1, a co-chaperone of Hsp90. Heat shock factor-1 was important for mediating the changes in Aha1 levels. Increased Aha1 levels were associated with enhanced interactions with Hsp90, resulting in increased Hsp90 ATPase activity. Moreover, increased Hsp90 ATPase activity resulted in increased phosphorylation of Akt and glycogen synthase kinase-3ß (GSK3ß), leading to enhanced expression of Wnt target genes. Significantly, levels of Aha1, Hsp90 ATPase activity, Akt, and GSK3ß phosphorylation and expression of Wnt target genes were increased in the colons of p53-null as compared with p53 wild type mice. Using p53 heterozygous mutant epithelial cells from Li-Fraumeni syndrome patients, we show that a monoallelic mutation of p53 was sufficient to activate the Aha1/Hsp90 ATPase axis leading to stimulation of Wnt signaling and increased expression of Wnt target genes. Pharmacologic intervention with CP-31398, a p53 rescue agent, inhibited recruitment of Aha1 to Hsp90 and suppressed Wnt-mediated gene expression in colon cancer cells. Taken together, this study provides new insights into the mechanism by which p53 regulates Wnt signaling and raises the intriguing possibility that p53 status may affect the efficacy of anticancer therapies targeting Hsp90 ATPase.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/genética , Proteína p53 Supresora de Tumor/metabolismo , Vía de Señalización Wnt/genética , Animales , Neoplasias del Colon/genética , Modelos Animales de Enfermedad , Humanos , Síndrome de Li-Fraumeni , Ratones , Ratones Transgénicos , Pirimidinas/farmacología , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA