Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gastroenterology ; 161(5): 1601-1614.e23, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34303658

RESUMEN

BACKGROUND & AIMS: Promoted by pancreatitis, oncogenic KrasG12D triggers acinar cells' neoplastic transformation through acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia. Anterior gradient 2 (Agr2), a known inhibitor of p53, is detected at early stage of pancreatic ductal adenocarcinoma (PDAC) development. RNA polymerase II (RNAPII) is a key nuclear enzyme; regulation of its nuclear localization in mammalian cells represents a potential therapeutic target. METHODS: A mouse model of inflammation-accelerated KrasG12D-driven ADM and pancreatic intraepithelial neoplasia development was used. Pancreas-specific Agr2 ablation was performed to access its role in pancreatic carcinogenesis. Hydrophobic hexapeptides loaded in liposomes were developed to disrupt Agr2-RNAPII complex. RESULTS: We found that Agr2 is up-regulated in ADM-to-pancreatic intraepithelial neoplasia transition in inflammation and KrasG12D-driven early pancreatic carcinogenesis. Genetic ablation of Agr2 specifically blocks this metaplastic-to-neoplastic process. Mechanistically, Agr2 directs the nuclear import of RNAPII via its C-terminal nuclear localization signal, undermining the ATR-dependent p53 activation in ADM lesions. Because Agr2 binds to the largest subunit of RNAPII in a peptide motif-dependent manner, we developed a hexapeptide to interfere with the nuclear import of RNAPII by competitively disrupting the Agr2-RNAPII complex. This novel hexapeptide leads to dysfunction of RNAPII with concomitant activation of DNA damage response in early neoplastic lesions; hence, it dramatically compromises PDAC initiation in vivo. Moreover, the hexapeptide sensitizes PDAC cells and patient-derived organoids harboring wild-type p53 to RNAPII inhibitors and first-line chemotherapeutic agents in vivo. Of note, this therapeutic effect is efficient across various cancer types. CONCLUSIONS: Agr2 is identified as a novel adaptor protein for nuclear import of RNAPII in mammalian cells. Also, we provide genetic evidence defining Agr2-dependent nuclear import of RNAPII as a pharmaceutically accessible target for prevention and treatment in PDAC in the context of wild-type p53.


Asunto(s)
Carcinoma in Situ/enzimología , Carcinoma Ductal Pancreático/enzimología , Mucoproteínas/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/enzimología , ARN Polimerasa II/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Transporte Activo de Núcleo Celular , Animales , Antineoplásicos/farmacología , Carcinoma in Situ/tratamiento farmacológico , Carcinoma in Situ/genética , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Metaplasia , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Mucoproteínas/genética , Mutación , Oligopéptidos/farmacología , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Polimerasa II/genética , Proteína p53 Supresora de Tumor/genética
2.
EMBO Rep ; 22(5): e51415, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33786993

RESUMEN

The tumour suppressors RNF43 and ZNRF3 play a central role in development and tissue homeostasis by promoting the turnover of the Wnt receptors LRP6 and Frizzled (FZD). The stem cell growth factor R-spondin induces auto-ubiquitination and membrane clearance of ZNRF3/RNF43 to promote Wnt signalling. However, the deubiquitinase stabilising ZNRF3/RNF43 at the plasma membrane remains unknown. Here, we show that the USP42 antagonises R-spondin by protecting ZNRF3/RNF43 from ubiquitin-dependent clearance. USP42 binds to the Dishevelled interacting region (DIR) of ZNRF3 and stalls the R-spondin-LGR4-ZNRF3 ternary complex by deubiquitinating ZNRF3. Accordingly, USP42 increases the turnover of LRP6 and Frizzled (FZD) receptors and inhibits Wnt signalling. Furthermore, we show that USP42 functions as a roadblock for paracrine Wnt signalling in colon cancer cells and mouse small intestinal organoids. We provide new mechanistic insights into the regulation R-spondin and conclude that USP42 is crucial for ZNRF3/RNF43 stabilisation at the cell surface.


Asunto(s)
Trombospondinas , Ubiquitina-Proteína Ligasas , Animales , Ratones , Receptores Acoplados a Proteínas G/genética , Trombospondinas/genética , Trombospondinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Vía de Señalización Wnt
3.
Mol Cell ; 81(6): 1170-1186.e10, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571422

RESUMEN

The complex architecture of transmembrane proteins requires quality control (QC) of folding, membrane positioning, and trafficking as prerequisites for cellular homeostasis and intercellular communication. However, it has remained unclear whether transmembrane protein-specific QC hubs exist. Here we identify cereblon (CRBN), the target of immunomodulatory drugs (IMiDs), as a co-chaperone that specifically determines chaperone activity of HSP90 toward transmembrane proteins by means of counteracting AHA1. This function is abrogated by IMiDs, which disrupt the interaction of CRBN with HSP90. Among the multiple transmembrane protein clients of CRBN-AHA1-HSP90 revealed by cell surface proteomics, we identify the amino acid transporter LAT1/CD98hc as a determinant of IMiD activity in multiple myeloma (MM) and present an Anticalin-based CD98hc radiopharmaceutical for MM radio-theranostics. These data establish the CRBN-AHA1-HSP90 axis in the biogenesis of transmembrane proteins, link IMiD activity to tumor metabolism, and nominate CD98hc and LAT1 as attractive diagnostic and therapeutic targets in MM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Factores Inmunológicos/farmacología , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Chaperonas Moleculares/metabolismo , Mieloma Múltiple/metabolismo , Proteínas de Neoplasias/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Células Tumorales Cultivadas
4.
J Clin Invest ; 130(6): 3270-3286, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32191641

RESUMEN

Single-nucleotide polymorphisms and locus amplification link the NF-κB transcription factor c-Rel to human autoimmune diseases and B cell lymphomas, respectively. However, the functional consequences of enhanced c-Rel levels remain enigmatic. Here, we overexpressed c-Rel specifically in mouse B cells from BAC-transgenic gene loci and demonstrate that c-Rel protein levels linearly dictated expansion of germinal center B (GCB) cells and isotype-switched plasma cells. c-Rel expression in B cells of otherwise c-Rel-deficient mice fully rescued terminal B cell differentiation, underscoring its critical B cell-intrinsic roles. Unexpectedly, in GCB cells transcription-independent regulation produced the highest c-Rel protein levels among B cell subsets. In c-Rel-overexpressing GCB cells this caused enhanced nuclear translocation, a profoundly altered transcriptional program, and increased proliferation. Finally, we provide a link between c-Rel gain and autoimmunity by showing that c-Rel overexpression in B cells caused autoantibody production and renal immune complex deposition.


Asunto(s)
Formación de Anticuerpos , Autoanticuerpos/inmunología , Centro Germinal/inmunología , Células Plasmáticas/inmunología , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-rel/inmunología , Animales , Autoanticuerpos/genética , Centro Germinal/patología , Ratones , Ratones Transgénicos , Células Plasmáticas/patología , Proteínas Proto-Oncogénicas c-rel/genética
5.
Nat Commun ; 11(1): 1268, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32152317

RESUMEN

Regulation of mitosis secures cellular integrity and its failure critically contributes to the development, maintenance, and treatment resistance of cancer. In yeast, the dual phosphatase Cdc14 controls mitotic progression by antagonizing Cdk1-mediated protein phosphorylation. By contrast, specific mitotic functions of the mammalian Cdc14 orthologue CDC14B have remained largely elusive. Here, we find that CDC14B antagonizes CDK1-mediated activating mitotic phosphorylation of the deubiquitinase USP9X at serine residue 2563, which we show to be essential for USP9X to mediate mitotic survival. Starting from an unbiased proteome-wide screening approach, we specify Wilms' tumor protein 1 (WT1) as the relevant substrate that becomes deubiquitylated and stabilized by serine 2563-phosphorylated USP9X in mitosis. We further demonstrate that WT1 functions as a mitotic transcription factor and specify CXCL8/IL-8 as a target gene of WT1 that conveys mitotic survival. Together, we describe a ubiquitin-dependent signaling pathway that directs a mitosis-specific transcription program to regulate mitotic survival.


Asunto(s)
Proteína Quinasa CDC2/antagonistas & inhibidores , Fosfatasas de Especificidad Dual/antagonistas & inhibidores , Mitosis/fisiología , Ubiquitina Tiolesterasa/efectos de los fármacos , Ubiquitina Tiolesterasa/metabolismo , Proteínas WT1/metabolismo , Células A549 , Apoptosis , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Interleucina-8/metabolismo , Fosforilación , Factores de Transcripción , Ubiquitina Tiolesterasa/genética , Proteínas WT1/genética
6.
Int Rev Cell Mol Biol ; 343: 219-297, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30712673

RESUMEN

Multiple myeloma (MM) is the second most common hematological malignancy and results from the clonal amplification of plasma cells. Despite recent advances in treatment, MM remains incurable with a median survival time of only 5-6years, thus necessitating further insights into MM biology and exploitation of novel therapeutic approaches. Both the ubiquitin proteasome system (UPS) and the PI3K/Akt/mTOR signaling pathways have been implicated in the pathogenesis, and treatment of MM and different lines of evidence suggest a close cross talk between these central cell-regulatory signaling networks. In this review, we outline the interplay between the UPS and mTOR pathways and discuss their implications for the pathophysiology and therapy of MM.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Ubiquitina/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/química , Inhibidores Enzimáticos/química , Humanos , Mieloma Múltiple/metabolismo , Sirolimus/química , Ubiquitina/metabolismo
7.
Nat Med ; 22(7): 735-43, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27294876

RESUMEN

Immunomodulatory drugs (IMiDs), such as thalidomide and its derivatives lenalidomide and pomalidomide, are key treatment modalities for hematologic malignancies, particularly multiple myeloma (MM) and del(5q) myelodysplastic syndrome (MDS). Cereblon (CRBN), a substrate receptor of the CRL4 ubiquitin ligase complex, is the primary target by which IMiDs mediate anticancer and teratogenic effects. Here we identify a ubiquitin-independent physiological chaperone-like function of CRBN that promotes maturation of the basigin (BSG; also known as CD147) and solute carrier family 16 member 1 (SLC16A1; also known as MCT1) proteins. This process allows for the formation and activation of the CD147-MCT1 transmembrane complex, which promotes various biological functions, including angiogenesis, proliferation, invasion and lactate export. We found that IMiDs outcompete CRBN for binding to CD147 and MCT1, leading to destabilization of the CD147-MCT1 complex. Accordingly, IMiD-sensitive MM cells lose CD147 and MCT1 expression after being exposed to IMiDs, whereas IMiD-resistant cells retain their expression. Furthermore, del(5q) MDS cells have elevated CD147 expression, which is attenuated after IMiD treatment. Finally, we show that BSG (CD147) knockdown phenocopies the teratogenic effects of thalidomide exposure in zebrafish. These findings provide a common mechanistic framework to explain both the teratogenic and pleiotropic antitumor effects of IMiDs.


Asunto(s)
Basigina/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Factores Inmunológicos/farmacología , Inmunosupresores/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Péptido Hidrolasas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Teratogénesis/efectos de los fármacos , Talidomida/farmacología , Proteínas Adaptadoras Transductoras de Señales , Basigina/genética , Basigina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos , Lenalidomida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Teratogénesis/genética , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligasas
8.
EMBO Mol Med ; 8(8): 851-62, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27317434

RESUMEN

The mitotic spindle assembly checkpoint (SAC) maintains genome stability and marks an important target for antineoplastic therapies. However, it has remained unclear how cells execute cell fate decisions under conditions of SAC-induced mitotic arrest. Here, we identify USP9X as the mitotic deubiquitinase of the X-linked inhibitor of apoptosis protein (XIAP) and demonstrate that deubiquitylation and stabilization of XIAP by USP9X lead to increased resistance toward mitotic spindle poisons. We find that primary human aggressive B-cell lymphoma samples exhibit high USP9X expression that correlate with XIAP overexpression. We show that high USP9X/XIAP expression is associated with shorter event-free survival in patients treated with spindle poison-containing chemotherapy. Accordingly, aggressive B-cell lymphoma lines with USP9X and associated XIAP overexpression exhibit increased chemoresistance, reversed by specific inhibition of either USP9X or XIAP. Moreover, knockdown of USP9X or XIAP significantly delays lymphoma development and increases sensitivity to spindle poisons in a murine Eµ-Myc lymphoma model. Together, we specify the USP9X-XIAP axis as a regulator of the mitotic cell fate decision and propose that USP9X and XIAP are potential prognostic biomarkers and therapeutic targets in aggressive B-cell lymphoma.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos B/efectos de los fármacos , Muerte Celular , Resistencia a Medicamentos , Linfoma de Células B/patología , Ubiquitina Tiolesterasa/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Animales , Linfocitos B/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Mitosis , Procesamiento Proteico-Postraduccional , Ubiquitina/metabolismo
9.
Nat Med ; 20(12): 1401-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25419709

RESUMEN

We searched for genetic alterations in human B cell lymphoma that affect the ubiquitin-proteasome system. This approach identified FBXO25 within a minimal common region of frequent deletion in mantle cell lymphoma (MCL). FBXO25 encodes an orphan F-box protein that determines the substrate specificity of the SCF (SKP1-CUL1-F-box)(FBXO25) ubiquitin ligase complex. An unbiased screen uncovered the prosurvival protein HCLS1-associated protein X-1 (HAX-1) as the bona fide substrate of FBXO25 that is targeted after apoptotic stresses. Protein kinase Cδ (PRKCD) initiates this process by phosphorylating FBXO25 and HAX-1, thereby spatially directing nuclear FBXO25 to mitochondrial HAX-1. Our analyses in primary human MCL identify monoallelic loss of FBXO25 and stabilizing HAX1 phosphodegron mutations. Accordingly, FBXO25 re-expression in FBXO25-deleted MCL cells promotes cell death, whereas expression of the HAX-1 phosphodegron mutant inhibits apoptosis. In addition, knockdown of FBXO25 significantly accelerated lymphoma development in Eµ-Myc mice and in a human MCL xenotransplant model. Together we identify a PRKCD-dependent proapoptotic mechanism controlling HAX-1 stability, and we propose that FBXO25 functions as a haploinsufficient tumor suppressor and that HAX1 is a proto-oncogene in MCL.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/genética , Proteínas F-Box/genética , Linfoma de Células B/genética , Linfoma de Células del Manto/genética , Proteínas del Tejido Nervioso/genética , Proteína Quinasa C-delta/genética , Proto-Oncogenes/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Animales , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células del Manto/metabolismo , Ratones , Proto-Oncogenes Mas , Transducción de Señal/genética
10.
Nat Cell Biol ; 15(1): 72-81, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23263282

RESUMEN

The Tel2 (also known as Telo2) and Tti1 proteins control the cellular abundance of mammalian PIKKs and are integral components of mTORC1 and mTORC2. Here we report that Tel2 and Tti1 are targeted for degradation within mTORC1 by the SCFFbxo9 ubiquitin ligase to adjust mTOR signalling to growth factor availability. This process is primed by CK2, which translocates to the cytoplasm to mediate mTORC1-specific phosphorylation of Tel2/Tti1, subsequent to growth factor deprivation. As a consequence, mTORC1 is inactivated to restrain cell growth and protein translation whereas relief of feedback inhibition activates the PI(3)K/TORC2/Akt pathway to sustain survival. Significantly, primary human multiple myelomas exhibit high levels of Fbxo9. In this setting, PI(3)K/TORC2/Akt signalling and survival of multiple myeloma cells is dependent on Fbxo9 expression. Thus, mTORC1-specific degradation of the Tel2 and Tti1 proteins represents a central mTOR regulatory mechanism with implications in multiple myeloma, both in promoting survival and in providing targets for the specific treatment of multiple myeloma with high levels of Fbxo9 expression.


Asunto(s)
Proteínas Portadoras/metabolismo , Quinasa de la Caseína II/fisiología , Supervivencia Celular , Proteínas F-Box/fisiología , Mieloma Múltiple/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-ets/metabolismo , Secuencia de Aminoácidos , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Medio de Cultivo Libre de Suero , Supervivencia sin Enfermedad , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Péptidos y Proteínas de Señalización Intracelular , Estimación de Kaplan-Meier , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mieloma Múltiple/patología , Complejos Multiproteicos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosforilación , Células Plasmáticas/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
11.
Mol Cell Biol ; 31(7): 1528-39, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21282470

RESUMEN

The chaperone-related AAA ATPase Cdc48 (p97/VCP in higher eukaryotes) segregates ubiquitylated proteins for subsequent degradation by the 26S proteasome or for nonproteolytic fates. The specific outcome of Cdc48 activity is controlled by the evolutionary conserved cofactors Ufd2 and Ufd3, which antagonistically regulate the substrates' ubiquitylation states. In contrast to the interaction of Ufd3 and Cdc48, the interaction between the ubiquitin chain elongating enzyme Ufd2 and Cdc48 has not been precisely mapped. Consequently, it is still unknown whether physiological functions of Ufd2 in fact require Cdc48 binding. Here, we show that Ufd2 binds to the C-terminal tail of Cdc48, unlike the human Ufd2 homologue E4B, which interacts with the N domain of p97. The binding sites for Ufd2 and Ufd3 on Cdc48 overlap and depend critically on the conserved residue Y834 but are not identical. Saccharomyces cerevisiae cdc48 mutants altered in residue Y834 or lacking the C-terminal tail are viable and exhibit normal growth. Importantly, however, loss of Ufd2 and Ufd3 binding in these mutants phenocopies defects of Δufd2 and Δufd3 mutants in the ubiquitin fusion degradation (UFD) and Ole1 fatty acid desaturase activation (OLE) pathways. These results indicate that key cellular functions of Ufd2 and Ufd3 in proteasomal protein degradation require their interaction with Cdc48.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Adenosina Trifosfatasas/química , Animales , Proteínas de Ciclo Celular/química , Secuencia Conservada/genética , Humanos , Mamíferos , Viabilidad Microbiana , Mutación/genética , Fosforilación , Unión Proteica , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/crecimiento & desarrollo , Relación Estructura-Actividad , Tirosina/metabolismo , Proteína que Contiene Valosina
12.
EMBO Rep ; 11(6): 479-85, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20414249

RESUMEN

The ubiquitin-selective chaperone p97 is involved in major proteolytic pathways of eukaryotic cells and has been implicated in several human proteinopathies. Moreover, mutations in p97 cause the disorder inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD). The molecular basis underlying impaired degradation and pathological aggregation of ubiquitinated proteins in IBMPFD is unknown. Here, we identify perturbed co-factor binding as a common defect of IBMPFD-causing mutant p97. We show that IBMPFD mutations induce conformational changes in the p97 N domain, the main binding site for regulatory co-factors. Consistently, mutant p97 proteins exhibit strongly altered co-factor interactions. Specifically, binding of the ubiquitin ligase E4B is reduced, whereas binding of ataxin 3 is enhanced, thus resembling the accumulation of mutant ataxin 3 on p97 in spinocerebellar ataxia type 3. Our results suggest that imbalanced co-factor binding to p97 is a key pathological feature of IBMPFD and potentially of other proteinopathies involving p97.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Demencia Frontotemporal/complicaciones , Demencia Frontotemporal/metabolismo , Miositis por Cuerpos de Inclusión/complicaciones , Miositis por Cuerpos de Inclusión/metabolismo , Osteítis Deformante/complicaciones , Osteítis Deformante/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfato/farmacología , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Demencia Frontotemporal/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación/genética , Proteínas Nucleares/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Proteínas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina-Proteína Ligasas , Proteína que Contiene Valosina
13.
Biochem Biophys Res Commun ; 380(2): 303-7, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19174149

RESUMEN

The chaperone-related p97 protein plays a central role in various cellular processes involving the ubiquitin-proteasome system. The diverse functions of p97 are controlled by a large number of cofactors that recruit specific substrates or influence their ubiquitylation state. Many cofactors bind through a UBX or PUB domain, two major p97 binding modules. However, the recently identified UBXD1 cofactor possesses both domains. To elucidate the molecular basis underlying the interaction between UBXD1 and p97, we analyzed the contribution of both domains to p97 binding biochemically and in living cells. The PUB domain mediated robust binding to the carboxy-terminus of p97, while the UBX domain did not contribute to p97 binding. Importantly, we identified an additional p97 binding site in UBXD1 that competed with the p47 cofactor for binding to the N domain of p97. This novel, bipartite binding mode suggests that UBXD1 could be an efficient regulator of p97 cofactor interactions.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Adenosina Trifosfatasas/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Relacionadas con la Autofagia , Sitios de Unión , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Humanos , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteína que Contiene Valosina
14.
J Biol Chem ; 283(3): 1381-1390, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17984091

RESUMEN

Thermal stress might lead to protein aggregation in the cell. Reactivation of protein aggregates depends on Hsp100 and Hsp70 chaperones. We focus in this study on the ability of DnaK, the bacterial representative of the Hsp70 family, to interact with different aggregated model substrates. Our data indicate that DnaK binding to large protein aggregates is mediated by DnaJ, and therefore it depends on its affinity for the cochaperone. Mutations in the structural region of DnaK known as the "latch" decrease the affinity of the chaperone for DnaJ, resulting in a defective activity as protein aggregate-removing agent. As expected, the chaperone activity is recovered when DnaJ concentration is raised to overcome the lower affinity of the mutant for the cochaperone, suggesting that a minimum number of aggregate-bound DnaK molecules is necessary for its efficient reactivation. Our results provide the first experimental evidence of DnaJ-mediated recruiting of ATP-DnaK molecules to the aggregate surface.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Escherichia coli/enzimología , Proteínas de Escherichia coli/química , Glucosafosfato Deshidrogenasa/metabolismo , Proteínas HSP70 de Choque Térmico/química , Cinética , Luciferasas/metabolismo , Malato Deshidrogenasa/metabolismo , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Proteínas Mutantes/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
15.
J Biol Chem ; 281(11): 7479-88, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16415343

RESUMEN

To gain further insight into the interactions involved in the allosteric transition of DnaK we have characterized wild-type (wt) protein and three mutants in which ionic interactions at the interface between the two subdomains of the substrate binding domain, and within the lid subdomain have been disrupted. Our data show that ionic contacts, most likely forming an electrically charged network, between the N-terminal region of helix B and an inner loop of the beta-sandwich are involved in maintaining the position of the lid relative to the beta-subdomain in the ADP state but not in the ATP state of the protein. Disruption of the ionic interactions between the C-terminal region of helix B and the outer loops of the beta-sandwich, known as the latch, does not have the same conformational consequences but results equally in an inactive protein. This indicates that a variety of mechanisms can inactivate this complex allosteric machine. Our results identify the ionic contacts at the subdomain and interdomain interfaces that are part of the hinge region involved in the ATP-induced allosteric displacement of the lid away from the peptide binding site. These interactions also stabilize peptide-Hsp70 complexes at physiological (37 degrees C) and stress (42 degrees C) temperatures, a requirement for productive substrate (re)folding.


Asunto(s)
Adenosina Trifosfatasas/química , Proteínas Bacterianas/química , Escherichia coli/metabolismo , Chaperonas Moleculares/química , Adenosina Difosfato/química , Adenosina Trifosfato/química , Regulación Alostérica , Sitio Alostérico , Secuencia de Aminoácidos , Anisotropía , Sitios de Unión , Rastreo Diferencial de Calorimetría , Calor , Iones , Cinética , Luciferasas/metabolismo , Espectrometría de Masas , Microscopía Fluorescente , Conformación Molecular , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Péptidos/química , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato , Temperatura , Termodinámica , Factores de Tiempo , Tripsina/química , Tripsina/farmacología
16.
Protein Sci ; 15(2): 223-33, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16384998

RESUMEN

The biological activity of DnaK, the bacterial representative of the Hsp70 protein family, is regulated by the allosteric interaction between its nucleotide and peptide substrate binding domains. Despite the importance of the nucleotide-induced cycling of DnaK between substrate-accepting and releasing states, the heterotropic allosteric mechanism remains as yet undefined. To further characterize this mechanism, the nucleotide-induced absorbance changes in the vibrational spectrum of wild-type DnaK was characterized. To assign the conformation sensitive absorption bands, two deletion mutants (one lacking the C-terminal alpha-helical subdomain and another comprising only the N-terminal ATPase domain), and a single-point DnaK mutant (T199A) with strongly reduced ATPase activity, were investigated by time-resolved infrared difference spectroscopy combined with the use of caged-nucleotides. The results indicate that (1) ATP, but not ADP, binding promotes a conformational change in both subdomains of the peptide binding domain that can be individually resolved; (2) these conformational changes are kinetically coupled, most likely to ensure a decrease in the affinity of DnaK for peptide substrates and a concomitant displacement of the lid away from the peptide binding site that would promote efficient diffusion of the released peptide to the medium; and (3) the alpha-helical subdomain contributes to stabilize the interdomain interface against the thermal challenge and allows bidirectional transmission of the allosteric signal between the ATPase and substrate binding domains at stress temperatures (42 degrees C).


Asunto(s)
Adenosina Difosfato/farmacología , Adenosina Trifosfato/farmacología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Regulación Alostérica , Escherichia coli , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Eliminación de Secuencia , Espectrofotometría Infrarroja , Especificidad por Sustrato
17.
FEBS J ; 272(12): 3184-96, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15955075

RESUMEN

Among the eukaryotic members of the Hsp70 family, mitochondrial Hsp70 shows the highest degree of sequence identity with bacterial DnaK. Although they share a functional mechanism and homologous co-chaperones, they are highly specific and cannot be exchanged between Escherichia coli and yeast mitochondria. To provide a structural basis for this finding, we characterized both proteins, as well as two DnaK/mtHsp70 chimeras constructed by domain swapping, using biochemical and biophysical methods. Here, we show that DnaK and mtHsp70 display different conformational and biochemical properties. Replacing different regions of the DnaK peptide-binding domain with those of mtHsp70 results in chimeric proteins that: (a) are not able to support growth of an E. coli DnaK deletion strain at stress temperatures (e.g. 42 degrees C); (b) show increased accessibility and decreased thermal stability of the peptide-binding pocket; and (c) have reduced activation by bacterial, but not mitochondrial co-chaperones, as compared with DnaK. Importantly, swapping the C-terminal alpha-helical subdomain promotes a conformational change in the chimeras to an mtHsp70-like conformation. Thus, interaction with bacterial co-chaperones correlates well with the conformation that natural and chimeric Hsp70s adopt in solution. Our results support the hypothesis that a specific protein structure might regulate the interaction of Hsp70s with particular components of the cellular machinery, such as Tim44, so that they perform specific functions.


Asunto(s)
Adenosina Trifosfatasas/química , Proteínas Bacterianas/química , Proteínas HSP70 de Choque Térmico/química , Mitocondrias/metabolismo , Chaperonas Moleculares/química , Levaduras/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Escherichia coli/genética , Escherichia coli/metabolismo , Prueba de Complementación Genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Fluorescencia , Espectrofotometría Infrarroja , Levaduras/genética
18.
J Biol Chem ; 279(19): 19600-6, 2004 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-14985342

RESUMEN

We examined the effect of deletion of different segments in the helical subdomain (the so-called "lid") of the DnaK peptide-binding domain on peptide binding and protein stability. At 25 degrees C, wt DnaK and the deletion mutant proteins are able to stably bind peptides with similar affinity. However, at physiological (37 degrees C) and stress (42 degrees C) temperatures, removal of the N-terminal half of alphaB and the rest of the lid drastically decreases the ability of the protein to bind substrates. Differential scanning calorimetry and infrared spectroscopy show that this behavior is accompanied by destabilization of the peptide-binding domain. Our data suggest that the reversible interaction between the lid and beta-sandwich subdomains of DnaK peptide-binding domain is required for the stabilization of the loops that form the peptide-binding site, which in turn modulates the protein affinity for peptide substrates. This interaction might have functional implications because it could prevent rebinding of the peptide substrate, which would be forced to fold.


Asunto(s)
Proteínas Bacterianas/química , Sitios de Unión , Rastreo Diferencial de Calorimetría , Eliminación de Gen , Rayos Infrarrojos , Cinética , Modelos Moleculares , Mutación , Péptidos/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Espectrofotometría , Espectrofotometría Infrarroja , Temperatura , Termodinámica , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...