Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(16): e2316651121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38588418

RESUMEN

Protecting chromosome ends from misrecognition as double-stranded (ds) DNA breaks is fundamental to eukaryotic viability. The protein complex shelterin prevents a DNA damage response at mammalian telomeres. Mammalian shelterin proteins TRF1 and TRF2 and their homologs in yeast and protozoa protect telomeric dsDNA. N-terminal homodimerization and C-terminal Myb-domain-mediated dsDNA binding are two structural hallmarks of end protection by TRF homologs. Yet our understanding of how Caenorhabditis elegans protects its telomeric dsDNA is limited. Recently identified C. elegans proteins TEBP-1 (also called DTN-1) and TEBP-2 (also called DTN-2) are functional homologs of TRF proteins, but how they bind DNA and whether or how they dimerize is not known. TEBP-1 and TEBP-2 harbor three Myb-containing domains (MCDs) and no obvious dimerization domain. We demonstrate biochemically that only the third MCD binds DNA. We solve the X-ray crystal structure of TEBP-2 MCD3 with telomeric dsDNA to reveal the structural mechanism of telomeric dsDNA protection in C. elegans. Mutagenesis of the DNA-binding site of TEBP-1 and TEBP-2 compromises DNA binding in vitro, and increases DNA damage signaling, lengthens telomeres, and decreases brood size in vivo. Via an X-ray crystal structure, biochemical validation of the dimerization interface, and SEC-MALS analysis, we demonstrate that MCD1 and MCD2 form a composite dimerization module that facilitates not only TEBP-1 and TEBP-2 homodimerization but also heterodimerization. These findings provide fundamental insights into C. elegans telomeric dsDNA protection and highlight how different eukaryotes have evolved distinct strategies to solve the chromosome end protection problem.


Asunto(s)
Proteínas de Caenorhabditis elegans , Proteínas de Unión a Telómeros , Animales , Proteínas de Unión a Telómeros/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Dimerización , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/química , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Unión Proteica , Telómero/genética , Telómero/metabolismo , Complejo Shelterina , ADN/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas , Mamíferos/genética
2.
Blood ; 138(10): 898-911, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34019641

RESUMEN

Germline pathogenic TERT variants are associated with short telomeres and an increased risk of developing myelodysplastic syndrome (MDS) among patients with a telomere biology disorder. We identified TERT rare variants in 41 of 1514 MDS patients (2.7%) without a clinical diagnosis of a telomere biology disorder who underwent allogeneic transplantation. Patients with a TERT rare variant had shorter telomere length (P < .001) and younger age at MDS diagnosis (52 vs 59 years, P = .03) than patients without a TERT rare variant. In multivariable models, TERT rare variants were associated with inferior overall survival (P = .034) driven by an increased incidence of nonrelapse mortality (NRM; P = .015). Death from a noninfectious pulmonary cause was more frequent among patients with a TERT rare variant. Most variants were missense substitutions and classified as variants of unknown significance. Therefore, we cloned all rare missense variants and quantified their impact on telomere elongation in a cell-based assay. We found that 90% of TERT rare variants had severe or intermediate impairment in their capacity to elongate telomeres. Using a homology model of human TERT bound to the shelterin protein TPP1, we inferred that TERT rare variants disrupt domain-specific functions, including catalysis, protein-RNA interactions, and recruitment to telomeres. Our results indicate that the contribution of TERT rare variants to MDS pathogenesis and NRM risk is underrecognized. Routine screening for TERT rare variants in MDS patients regardless of age or clinical suspicion may identify clinically inapparent telomere biology disorders and improve transplant outcomes through risk-adapted approaches.


Asunto(s)
Variación Genética , Síndromes Mielodisplásicos , Telomerasa/genética , Adulto , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Síndromes Mielodisplásicos/enzimología , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/mortalidad , Tasa de Supervivencia
3.
Proc Natl Acad Sci U S A ; 116(52): 26505-26515, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31822618

RESUMEN

Telomerase catalyzes telomeric DNA synthesis at chromosome ends to allow for continued cell division. The telomeric protein TPP1 is essential for enhancing the processivity of telomerase and recruiting the enzyme to telomeres. The telomerase interaction surface on human TPP1 has been mapped to 2 regions of the N-terminal oligosaccharide/oligonucleotide-binding (OB) domain, namely the TPP1 glutamate (E) and leucine (L)-rich (TEL) patch and the N terminus of TPP1-oligosaccharide/oligonucleotide-binding (NOB) region. To map the telomerase side of the interface, we exploited the predicted structural similarities for human and Tetrahymena thermophila telomerase as well as the species specificity of human and mouse telomerase for their cognate TPP1 partners. We show that swapping in the telomerase essential N-terminal (TEN) and insertions in fingers domain (IFD)-TRAP regions of the human telomerase catalytic protein subunit TERT into the mouse TERT backbone is sufficient to bias the species specificity toward human TPP1. Employing a structural homology-based mutagenesis screen focused on surface residues of the TEN and IFD regions, we identified TERT residues that are critical for contacting TPP1 but dispensable for other aspects of telomerase structure or function. We present a functionally validated structural model for how human telomerase engages TPP1 at telomeres, setting the stage for a high-resolution structure of this interface.

4.
Proc Natl Acad Sci U S A ; 113(46): 13021-13026, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807141

RESUMEN

Telomerase replicates chromosome ends to facilitate continued cell division. Mutations that compromise telomerase function result in stem cell failure diseases, such as dyskeratosis congenita (DC). One such mutation (K170Δ), residing in the telomerase-recruitment factor TPP1, provides an excellent opportunity to structurally, biochemically, and genetically dissect the mechanism of such diseases. We show through site-directed mutagenesis and X-ray crystallography that this TPP1 disease mutation deforms the conformation of two critical amino acids of the TEL [TPP1's glutamate (E) and leucine-rich (L)] patch, the surface of TPP1 that binds telomerase. Using CRISPR-Cas9 technology, we demonstrate that introduction of this mutation in a heterozygous manner is sufficient to shorten telomeres in human cells. Our findings rule out dominant-negative effects of the mutation. Instead, these findings implicate reduced TEL patch dosage in causing telomere shortening. Our studies provide mechanistic insight into telomerase-deficiency diseases and encourage the development of gene therapies to counter such diseases.


Asunto(s)
Disqueratosis Congénita/genética , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética , Línea Celular Tumoral , Cristalografía por Rayos X , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Conformación Proteica , Complejo Shelterina , Telomerasa/metabolismo , Telómero/metabolismo
5.
Proc Natl Acad Sci U S A ; 108(37): E667-73, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21876134

RESUMEN

Few experimental techniques can assess the orientation of peripheral membrane proteins in their native environment. Sum Frequency Generation (SFG) vibrational spectroscopy was applied to study the formation of the complex between G protein-coupled receptor (GPCR) kinase 2 (GRK2) and heterotrimeric G protein ß(1)γ(2) subunits (Gßγ) at a lipid bilayer, without any exogenous labels. The most likely membrane orientation of the GRK2-Gßγ complex differs from that predicted from the known protein crystal structure, and positions the predicted receptor docking site of GRK2 such that it would more optimally interact with GPCRs. Gßγ also appears to change its orientation after binding to GRK2. The developed methodology is widely applicable for the study of other membrane proteins in situ.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/química , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades beta de la Proteína de Unión al GTP/química , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/química , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Membranas Artificiales , Membrana Dobles de Lípidos/química , Modelos Moleculares , Fosfatidilcolinas/química , Fosfatidilgliceroles/química , Unión Proteica , Conformación Proteica , Análisis Espectral , Vibración
6.
Science ; 381(6659): 771-778, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37590346

RESUMEN

Protection of telomeres 1 (POT1) is the 3' single-stranded overhang-binding telomeric protein that prevents an ataxia telangiectasia and Rad3-related (ATR) DNA damage response (DDR) at chromosome ends. What precludes the DDR machinery from accessing the telomeric double-stranded-single-stranded junction is unknown. We demonstrate that human POT1 binds this junction by recognizing the phosphorylated 5' end of the chromosome. High-resolution crystallographic structures reveal that the junction is capped by POT1 through a "POT-hole" surface, the mutation of which compromises junction protection in vitro and telomeric 5'-end definition and DDR suppression in human cells. Whereas both mouse POT1 paralogs bind the single-stranded overhang, POT1a, not POT1b, contains a POT-hole and binds the junction, which explains POT1a's sufficiency for end protection. Our study shifts the paradigm for DDR suppression at telomeres by highlighting the importance of protecting the double-stranded-single-stranded junction.


Asunto(s)
ADN , Complejo Shelterina , Proteínas de Unión a Telómeros , Telómero , Animales , Humanos , Ratones , Cristalografía , ADN/química , ADN/metabolismo , Mutación , Complejo Shelterina/química , Complejo Shelterina/genética , Complejo Shelterina/metabolismo , Telómero/química , Telómero/metabolismo , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
7.
Life Sci Alliance ; 6(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36963832

RESUMEN

Telomerase processively adds telomeric DNA repeats to chromosome ends using catalytic protein subunit TERT and a template on its RNA subunit TR. Mammalian telomerase is recruited to telomeres by the TEL patch and NOB regions of shelterin component TPP1. Recent cryo-EM structures of human telomerase reveal that a composite TERT TEN-(IFD-TRAP) domain interacts with TPP1. Here, we generate TERT mutants to demonstrate that a three-way TEN-(IFD-TRAP)-TPP1 interaction is critical for telomerase recruitment to telomeres and processive telomere repeat addition. Single mutations of IFD-TRAP at its interface with TR or the DNA primer impair telomerase catalysis. We further reveal the importance of TERT motif 3N and TEN domain loop 99FGF101 in telomerase action. Finally, we demonstrate that TPP1 TEL patch loop residue F172, which undergoes a structural rearrangement to bind telomerase, contributes to the human-mouse species specificity of the telomerase-TPP1 interaction. Our study provides insights into the multiple functions of TERT IFD-TRAP, reveals novel TERT and TPP1 elements critical for function, and helps explain how TPP1 binding licenses robust telomerase action at natural chromosome ends.


Asunto(s)
Telomerasa , Animales , Humanos , Ratones , Telomerasa/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Complejo Shelterina , Telómero/genética , Telómero/metabolismo , Mutación , Mamíferos/genética
8.
Nat Commun ; 12(1): 5514, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34535663

RESUMEN

Human shelterin components POT1 and TPP1 form a stable heterodimer that protects telomere ends from ATR-dependent DNA damage responses and regulates telomerase-dependent telomere extension. Mice possess two functionally distinct POT1 proteins. POT1a represses ATR/CHK1 DNA damage responses and the alternative non-homologous end-joining DNA repair pathway while POT1b regulates C-strand resection and recruits the CTC1-STN1-TEN1 (CST) complex to telomeres to mediate C-strand fill-in synthesis. Whether POT1a and POT1b are involved in regulating the length of the telomeric G-strand is unclear. Here we demonstrate that POT1b, independent of its CST function, enhances recruitment of telomerase to telomeres through three amino acids in its TPP1 interacting C-terminus. POT1b thus coordinates the synthesis of both telomeric G- and C-strands. In contrast, POT1a negatively regulates telomere length by inhibiting telomerase recruitment to telomeres. The identification of unique amino acids between POT1a and POT1b helps us understand mechanistically how human POT1 switches between end protective functions and promoting telomerase recruitment.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Telomerasa/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Análisis Mutacional de ADN , Ratones , Unión Proteica , Recombinasa Rad51/metabolismo , Sarcoma/patología
9.
JCI Insight ; 6(9)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33822766

RESUMEN

Telomerase catalyzes chromosome end replication in stem cells and other long-lived cells. Mutations in telomerase or telomere-related genes result in diseases known as telomeropathies. Telomerase is recruited to chromosome ends by the ACD/TPP1 protein (TPP1 hereafter), a component of the shelterin complex that protects chromosome ends from unwanted end joining. TPP1 facilitates end protection by binding shelterin proteins POT1 and TIN2. TPP1 variants have been associated with telomeropathies but remain poorly characterized in vivo. Disease variants and mutagenesis scans provide efficient avenues to interrogate the distinct physiological roles of TPP1. Here, we conduct mutagenesis in the TIN2- and POT1-binding domains of TPP1 to discover mutations that dissect TPP1's functions. Our results extend current structural data to reveal that the TPP1-TIN2 interface is more extensive than previously thought and highlight the robustness of the POT1-TPP1 interface. Introduction of separation-of-function mutants alongside known TPP1 telomeropathy mutations in mouse hematopoietic stem cells (mHSCs) lacking endogenous TPP1 demonstrated a clear phenotypic demarcation. TIN2- and POT1-binding mutants were unable to rescue mHSC failure resulting from end deprotection. In contrast, TPP1 telomeropathy mutations sustained mHSC viability, consistent with their selectively impacting end replication. These results highlight the power of scanning mutagenesis in revealing structural interfaces and dissecting multifunctional genes.


Asunto(s)
Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Complejo Shelterina/metabolismo , Proteínas de Unión a Telómeros/genética , Animales , Supervivencia Celular/genética , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Complejo Shelterina/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
10.
Cell Rep ; 27(12): 3511-3521.e7, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216472

RESUMEN

Telomerase replicates chromosome ends in germ and somatic stem cells to facilitate their continued proliferation. Telomerase action depends on the telomeric protein TPP1, which recruits telomerase to telomeres and facilitates processive DNA synthesis. Here, we identify separation-of-function long (TPP1-L) and short (TPP1-S) isoforms of TPP1 that appear to be generated from separate transcripts and differ only in 86 amino acids at their N terminus. Although both isoforms retain the ability to recruit telomerase, only TPP1-S facilitates efficient telomere synthesis. We find that TPP1-S is the predominant isoform in somatic cells, and strikingly, TPP1-L is the major isoform in differentiated male germ cells. We observed that TERT expression persists in these germ cells, suggesting that TPP1-L could restrain telomerase in this context. We show how differential expression of TPP1 isoforms determines telomerase function and demonstrate how alternative transcription start sites allow one gene to perform distinct functions in different biological contexts.


Asunto(s)
Aminopeptidasas/metabolismo , Cromosomas Humanos/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Células Germinativas/metabolismo , Serina Proteasas/metabolismo , Complejo Shelterina , Telomerasa/metabolismo , Homeostasis del Telómero , Proteínas de Unión a Telómeros , Testículo/metabolismo , Secuencia de Aminoácidos , Aminopeptidasas/genética , Cromosomas Humanos/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Células Germinativas/citología , Células HeLa , Humanos , Masculino , Unión Proteica , Isoformas de Proteínas , Homología de Secuencia , Serina Proteasas/genética , Complejo Shelterina/genética , Complejo Shelterina/metabolismo , Telomerasa/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Testículo/citología
11.
Cell Rep ; 22(5): 1132-1140, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29386102

RESUMEN

Telomerase recruitment to telomeres and enzymatic processivity are mediated by TPP1, an essential component of telomere integrity and telomerase function. A surface on the OB domain of TPP1 called the TEL patch is critical for TPP1's telomerase-associated functions. Here, we identify a separate region in the N terminus of the OB domain (termed NOB) of TPP1 that, like the TEL patch, is essential for telomerase repeat addition processivity in vitro as well as telomerase recruitment to telomeres and telomere lengthening in cells. Although well-conserved among most mammalian TPP1 homologs, the NOB region in mice is distinct. Swapping the sequence of human NOB into mouse TPP1 allows it to stimulate human telomerase, qualifying NOB as an important determinant of species specificity for TPP1-telomerase interaction. Our studies show that TPP1 NOB is critical for telomerase function and demonstrate that the telomerase interaction surface on TPP1 is more elaborate than previously appreciated.


Asunto(s)
Telomerasa/metabolismo , Homeostasis del Telómero/fisiología , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Quimera , Humanos , Ratones , Modelos Moleculares , Dominios Proteicos , Complejo Shelterina
12.
Nat Struct Mol Biol ; 24(12): 1064-1072, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29083414

RESUMEN

Tethering telomeres to the inner nuclear membrane (INM) allows homologous chromosome pairing during meiosis. The meiosis-specific protein TERB1 binds the telomeric protein TRF1 to establish telomere-INM connectivity and is essential for mouse fertility. Here we solve the structure of the human TRF1-TERB1 interface to reveal the structural basis for telomere-INM linkage. Disruption of this interface abrogates binding and compromises telomere-INM attachment in mice. An embedded CDK-phosphorylation site within the TRF1-binding region of TERB1 provides a mechanism for cap exchange, a late-pachytene phenomenon involving the dissociation of the TRF1-TERB1 complex. Indeed, further strengthening this interaction interferes with cap exchange. Finally, our biochemical analysis implicates distinct complexes for telomere-INM tethering and chromosome-end protection during meiosis. Our studies unravel the structure, stoichiometry, and physiological implications underlying telomere-INM tethering, thereby providing unprecedented insights into the unique function of telomeres in meiosis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Meiosis/fisiología , Membrana Nuclear/metabolismo , Telómero/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Sitios de Unión/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Emparejamiento Cromosómico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Unión Proteica/fisiología , Proteínas de Unión al GTP rap1/metabolismo
13.
Biochim Biophys Acta ; 1577(1): 102-8, 2002 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12151100

RESUMEN

In monocyte/macrophages, the human NF-IL6 gene was activated by LPS or PMA. However, a robust response required stimulation of cells with both LPS and PMA. To examine the molecular basis of this response, we isolated human genomic DNA and determined the nucleotide sequence of a segment (6.4 kb) that included the transcription initiation site of the gene. The unique sequences in the 6.4-kb DNA include several potential transcription factor-binding elements that may explain the molecular basis of the activation of the human NF-IL6 gene by signaling molecules that control the immune and inflammatory responses. Deletion analysis localized an LPS+PMA responsive region downstream position -287, with respect to the transcription initiation site of the NF-IL6 gene. The responsive region includes a potential site for interactions with CREB and a region (-287 to -247) that interacts with SP1 and SP3. In functional assays, the potential CREB site responded to cellular stimulation. The region that interacted with SP1 and SP3 augmented the overall level of activity produced in response to LPS+PMA.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Macrófagos/inmunología , Monocitos/inmunología , Regiones Promotoras Genéticas , Activación Transcripcional , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Células Cultivadas , Humanos , Cinética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , ARN Mensajero/biosíntesis , Elementos de Respuesta , Factor de Transcripción Sp1/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/metabolismo , Regulación hacia Arriba
14.
Virus Res ; 89(1): 53-63, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12367750

RESUMEN

To examine the mechanism of HIV-1 regulation by NF-IL6 in activated human cells, we selected a Jurkat cell line that did not contain endogenous NF-IL6. In this cellular environment, we evaluated the effect of exogenous NF-IL6 on transcription mediated by native and deleted LTR sequences. In Jurkat cells stimulated with LPS and PMA, LTR-mediated transcription was enhanced by NF-IL6. The results of deletion studies revealed a central role for the basal LTR region and the TATA element in the LTR, in upregulation of reporter gene expression by NF-IL6 in activated cells. In the selected cellular environment, regulation of transcription by NF-IL6 was not evident in studies of promoter regions of other genes. The results implied that the basal region of HIV-1 LTR includes molecular properties that support activation of HIV-1 by NF-IL6 in stimulated cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/farmacología , Regulación Viral de la Expresión Génica , VIH-1/metabolismo , Células Jurkat/virología , Transcripción Genética/efectos de los fármacos , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Humanos , Activación de Linfocitos , Regiones Promotoras Genéticas
15.
Structure ; 21(3): 438-48, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23434405

RESUMEN

The heterotrimeric G protein Gαq is a key regulator of blood pressure, and excess Gαq signaling leads to hypertension. A specific inhibitor of Gαq is the GTPase activating protein (GAP) known as regulator of G protein signaling 2 (RGS2). The molecular basis for how Gαq/11 subunits serve as substrates for RGS proteins and how RGS2 mandates its selectivity for Gαq is poorly understood. In crystal structures of the RGS2-Gαq complex, RGS2 docks to Gαq in a different orientation from that observed in RGS-Gαi/o complexes. Despite its unique pose, RGS2 maintains canonical interactions with the switch regions of Gαq in part because its α6 helix adopts a distinct conformation. We show that RGS2 forms extensive interactions with the α-helical domain of Gαq that contribute to binding affinity and GAP potency. RGS subfamilies that do not serve as GAPs for Gαq are unlikely to form analogous stabilizing interactions.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Simulación de Dinámica Molecular , Subunidades de Proteína/química , Proteínas RGS/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bovinos , Cristalografía por Rayos X , Escherichia coli/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Humanos , Ratones , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Subunidades de Proteína/genética , Proteínas RGS/genética , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alineación de Secuencia , Relación Estructura-Actividad
16.
Structure ; 20(8): 1300-9, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22727813

RESUMEN

Cardiovascular homeostasis is maintained in part by the rapid desensitization of activated heptahelical receptors that have been phosphorylated by G protein-coupled receptor kinase 2 (GRK2). However, during chronic heart failure GRK2 is upregulated and believed to contribute to disease progression. We have determined crystallographic structures of GRK2 bound to an RNA aptamer that potently and selectively inhibits kinase activity. Key to the mechanism of inhibition is the positioning of an adenine nucleotide into the ATP-binding pocket and interactions with the basic αF-αG loop region of the GRK2 kinase domain. Constraints imposed on the RNA by the terminal stem of the aptamer also play a role. These results highlight how a high-affinity aptamer can be used to selectively trap a novel conformational state of a protein kinase.


Asunto(s)
Aptámeros de Nucleótidos/química , Quinasa 2 del Receptor Acoplado a Proteína-G/química , Secuencias de Aminoácidos , Animales , Secuencia de Bases , Dominio Catalítico , Bovinos , Cristalografía por Rayos X , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Enlace de Hidrógeno , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Estructura Cuaternaria de Proteína
17.
Nat Struct Mol Biol ; 18(9): 999-1005, 2011 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-21822282

RESUMEN

The enzyme phospholipase C-ß (PLCß) is a crucial regulator of intracellular calcium levels whose activity is controlled by heptahelical receptors that couple to members of the Gq family of heterotrimeric G proteins. We have determined atomic structures of two invertebrate homologs of PLCß (PLC21) from cephalopod retina and identified a helix from the C-terminal regulatory region that interacts with a conserved surface of the catalytic core of the enzyme. Mutations designed to disrupt the analogous interaction in human PLCß3 considerably increase basal activity and diminish stimulation by Gαq. Gαq binding requires displacement of the autoinhibitory helix from the catalytic core, thus providing an allosteric mechanism for activation of PLCß.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Loligo/enzimología , Fosfolipasa C beta/química , Sepia/enzimología , Animales , Cristalografía por Rayos X , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fosfolipasa C beta/fisiología , Estructura Secundaria de Proteína/fisiología , Estructura Terciaria de Proteína
18.
J Med Chem ; 53(4): 1867-70, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20128603

RESUMEN

G protein-coupled receptor kinase 2 (GRK2) is a pharmaceutical target for the treatment of cardiovascular diseases such as congestive heart failure, myocardial infarction, and hypertension. To better understand how nanomolar inhibition and selectivity for GRK2 might be achieved, we have determined crystal structures of human GRK2 in complex with Gbetagamma in the presence and absence of the AGC kinase inhibitor balanol. The selectivity of balanol among human GRKs is assessed.


Asunto(s)
Azepinas/química , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Quinasa 2 del Receptor Acoplado a Proteína-G/química , Subunidades beta de la Proteína de Unión al GTP/química , Subunidades gamma de la Proteína de Unión al GTP/química , Hidroxibenzoatos/química , Modelos Moleculares , Cristalografía por Rayos X , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Humanos , Fosforilación , Unión Proteica , Conformación Proteica , Tubulina (Proteína)/química
19.
J Biol Chem ; 283(50): 34923-34, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18936096

RESUMEN

Transmembrane signaling through G alpha(q)-coupled receptors is linked to physiological processes such as cardiovascular development and smooth muscle function. Recent crystallographic studies have shown how G alpha(q) interacts with two activation-dependent targets, p63RhoGEF and G protein-coupled receptor kinase 2 (GRK2). These proteins bind to the effector-binding site of G alpha(q) in a manner that does not appear to physically overlap with the site on G alpha(q) bound by regulator of G-protein signaling (RGS) proteins, which function as GTPase-activating proteins (GAPs). Herein we confirm the formation of RGS-G alpha(q)-GRK2/p63RhoGEF ternary complexes using flow cytometry protein interaction and GAP assays. RGS2 and, to a lesser extent, RGS4 are negative allosteric modulators of Galpha(q) binding to either p63RhoGEF or GRK2. Conversely, GRK2 enhances the GAP activity of RGS4 but has little effect on that of RGS2. Similar but smaller magnitude responses are induced by p63RhoGEF. The fact that GRK2 and p63RhoGEF respond similarly to these RGS proteins supports the hypothesis that GRK2 is a bona fide G alpha(q) effector. The results also suggest that signal transduction pathways initiated by GRK2, such as the phosphorylation of G protein-coupled receptors, and by p63RhoGEF, such as the activation of gene transcription, can be regulated by RGS proteins via both allosteric and GAP mechanisms.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas RGS/química , Sitio Alostérico , Unión Competitiva , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo/métodos , GTP Fosfohidrolasas/metabolismo , Humanos , Cinética , Conformación Molecular , Unión Proteica , Estructura Terciaria de Proteína , Factores de Intercambio de Guanina Nucleótido Rho
20.
J Biol Chem ; 281(24): 16785-93, 2006 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-16613860

RESUMEN

We describe the 2.6-A crystal structure of human G protein-coupled receptor kinase (GRK)-6, a key regulator of dopaminergic signaling and lymphocyte chemotaxis. GRK6 is a member of the GRK4 subfamily of GRKs, which is represented in most, if not all, metazoans. Comparison of GRK6 with GRK2 confirms that the catalytic core of all GRKs consists of intimately associated kinase and regulator of G protein signaling (RGS) homology domains. Despite being in complex with an ATP analog, the kinase domain of GRK6 remains in an open, presumably inactive conformation, suggesting that G protein-coupled receptors activate GRKs by inducing kinase domain closure. The structure reveals a putative phospholipid-binding site near the N terminus of GRK6 and structural elements within the kinase substrate channel that likely influence G protein-coupled receptor access and specificity. The crystalline GRK6 RGS homology domain forms an extensive dimer interface using conserved hydrophobic residues distinct from those in GRK2 that bind Galpha(q), although dimerization does not appear to occur in solution and is not required for receptor phosphorylation.


Asunto(s)
Proteínas Serina-Treonina Quinasas/fisiología , Adenosina Trifosfato/química , Animales , Sitios de Unión , Linaje de la Célula , Cristalografía por Rayos X , Dimerización , Quinasas de Receptores Acoplados a Proteína-G , Humanos , Insectos , Conformación Molecular , Fosforilación , Conformación Proteica , Proteínas Serina-Treonina Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA