Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 161(5): 1601-1614.e23, 2021 11.
Article in English | MEDLINE | ID: mdl-34303658

ABSTRACT

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.


Subject(s)
Carcinoma in Situ/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Mucoproteins/metabolism , Oncogene Proteins/metabolism , Pancreatic Neoplasms/enzymology , RNA Polymerase II/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Antineoplastic Agents/pharmacology , Carcinoma in Situ/drug therapy , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Metaplasia , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mucoproteins/genetics , Mutation , Oligopeptides/pharmacology , Oncogene Proteins/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , RNA Polymerase II/genetics , Tumor Suppressor Protein p53/genetics
2.
EMBO Rep ; 22(5): e51415, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33786993

ABSTRACT

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.


Subject(s)
Thrombospondins , Ubiquitin-Protein Ligases , Animals , Mice , Receptors, G-Protein-Coupled/genetics , Thrombospondins/genetics , Thrombospondins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Wnt Signaling Pathway
3.
Mol Cell ; 81(6): 1170-1186.e10, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33571422

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , HSP90 Heat-Shock Proteins/metabolism , Immunologic Factors/pharmacology , Large Neutral Amino Acid-Transporter 1/metabolism , Molecular Chaperones/metabolism , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Tumor Cells, Cultured
4.
J Clin Invest ; 130(6): 3270-3286, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32191641

ABSTRACT

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.


Subject(s)
Antibody Formation , Autoantibodies/immunology , Germinal Center/immunology , Plasma Cells/immunology , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-rel/immunology , Animals , Autoantibodies/genetics , Germinal Center/pathology , Mice , Mice, Transgenic , Plasma Cells/pathology , Proto-Oncogene Proteins c-rel/genetics
5.
Nat Commun ; 11(1): 1268, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152317

ABSTRACT

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.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Dual-Specificity Phosphatases/antagonists & inhibitors , Mitosis/physiology , Ubiquitin Thiolesterase/drug effects , Ubiquitin Thiolesterase/metabolism , WT1 Proteins/metabolism , A549 Cells , Apoptosis , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Interleukin-8/metabolism , Phosphorylation , Transcription Factors , Ubiquitin Thiolesterase/genetics , WT1 Proteins/genetics
7.
Int Rev Cell Mol Biol ; 343: 219-297, 2019.
Article in English | MEDLINE | ID: mdl-30712673

ABSTRACT

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.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Enzyme Inhibitors/pharmacology , Multiple Myeloma/drug therapy , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Ubiquitin/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/chemistry , Enzyme Inhibitors/chemistry , Humans , Multiple Myeloma/metabolism , Sirolimus/chemistry , Ubiquitin/metabolism
8.
Nat Med ; 22(7): 735-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27294876

ABSTRACT

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.


Subject(s)
Basigin/drug effects , Cell Cycle Proteins/drug effects , Immunologic Factors/pharmacology , Immunosuppressive Agents/pharmacology , Oncogene Proteins/drug effects , Peptide Hydrolases/drug effects , RNA, Messenger/drug effects , Teratogenesis/drug effects , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Basigin/genetics , Basigin/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Humans , Lenalidomide , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Teratogenesis/genetics , Thalidomide/analogs & derivatives , Ubiquitin-Protein Ligases
9.
EMBO Mol Med ; 8(8): 851-62, 2016 08.
Article in English | MEDLINE | ID: mdl-27317434

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Cell Death , Drug Resistance , Lymphoma, B-Cell/pathology , Ubiquitin Thiolesterase/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , B-Lymphocytes/physiology , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mitosis , Protein Processing, Post-Translational , Ubiquitin/metabolism
10.
Nat Med ; 20(12): 1401-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25419709

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis/genetics , F-Box Proteins/genetics , Lymphoma, B-Cell/genetics , Lymphoma, Mantle-Cell/genetics , Nerve Tissue Proteins/genetics , Protein Kinase C-delta/genetics , Proto-Oncogenes/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Animals , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, Mantle-Cell/metabolism , Mice , Proto-Oncogene Mas , Signal Transduction/genetics
11.
Nat Cell Biol ; 15(1): 72-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23263282

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Casein Kinase II/physiology , Cell Survival , F-Box Proteins/physiology , Multiple Myeloma/metabolism , Proteolysis , Proto-Oncogene Proteins c-ets/metabolism , Amino Acid Sequence , Animals , Case-Control Studies , Cell Line, Tumor , Culture Media, Serum-Free , Disease-Free Survival , F-Box Proteins/genetics , F-Box Proteins/metabolism , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/physiology , Intracellular Signaling Peptides and Proteins , Kaplan-Meier Estimate , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Multiple Myeloma/pathology , Multiprotein Complexes/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Plasma Cells/metabolism , Protein Binding , Protein Processing, Post-Translational , Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
12.
Mol Cell Biol ; 31(7): 1528-39, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21282470

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Adenosine Triphosphatases/chemistry , Animals , Cell Cycle Proteins/chemistry , Conserved Sequence/genetics , Humans , Mammals , Microbial Viability , Mutation/genetics , Phosphorylation , Protein Binding , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/growth & development , Structure-Activity Relationship , Tyrosine/metabolism , Valosin Containing Protein
13.
EMBO Rep ; 11(6): 479-85, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20414249

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Frontotemporal Dementia/complications , Frontotemporal Dementia/metabolism , Myositis, Inclusion Body/complications , Myositis, Inclusion Body/metabolism , Osteitis Deformans/complications , Osteitis Deformans/metabolism , Adaptor Proteins, Vesicular Transport , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line , Frontotemporal Dementia/genetics , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Nuclear Proteins/metabolism , Protein Binding/drug effects , Protein Structure, Secondary , Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases , Valosin Containing Protein
14.
Biochem Biophys Res Commun ; 380(2): 303-7, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19174149

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Molecular Chaperones/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Adenosine Triphosphatases/genetics , Amino Acid Motifs , Amino Acid Sequence , Autophagy-Related Proteins , Binding Sites , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Humans , Molecular Chaperones/genetics , Molecular Sequence Data , Protein Interaction Mapping , Protein Structure, Tertiary , Valosin Containing Protein
15.
Pharmacol Rep ; 60(3): 391-8, 2008.
Article in English | MEDLINE | ID: mdl-18622064

ABSTRACT

We have tested the hypothesis that a pharmacologically determined alteration in renal alpha(2)-adrenoceptor (alpha(2)-AR) density might be a pathophysiologically important factor of genetic hypertension in the spontaneously hypertensive rats (SHRs). First, we compared he regional distribution and biochemical parameters of alpha(2)-ARs in SHRs and Wistar-Kyoto (WKY) rats, using the full agonist [(3)H]UK 14304. Secondly, we evaluated the effect of selective blockade and stimulation of alpha(2)-ARs on the development of hypertension and on renal alpha(2)-AR density and regional distribution in SHRs. [(3)H]UK 14304 binding was distributed predominantly over the outer medulla, less abundantly over the inner medulla and was almost absent from the renal cortex. Renal alpha(2)-ARs were found to be increased in SHRs at the ages tested compared with their respective controls and the increase was completely localized to the outer medulla. In these rats, blood pressures immediately before sacrifice were significantly higher in the hypertensive group compared with normotensive controls. The daily administration of SK&F 86,466 or clonidine significantly decreased the blood pressure but the autoradiographic studies showed that the prolonged administration of yohimbine to rats for two weeks resulted in a large increase in the density of alpha(2)-ARs in some areas of the rat kidney but not in others. Taken together these data do not support the hypothesis that alteration in renal alpha(2)-ARs (as measured by autoradiography) is crucial for the maintenance of hypertension in the SHR model.


Subject(s)
Hypertension/physiopathology , Kidney Cortex/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Yohimbine/pharmacology , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Analysis of Variance , Animals , Autoradiography/methods , Benzazepines/pharmacology , Blood Pressure/drug effects , Brimonidine Tartrate , Clonidine/pharmacology , Heart Rate/drug effects , Kidney Cortex/metabolism , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Male , Quinoxalines/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Tritium
16.
J Biol Chem ; 283(3): 1381-1390, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-17984091

ABSTRACT

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.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Glucosephosphate Dehydrogenase/metabolism , HSP70 Heat-Shock Proteins/chemistry , Kinetics , Luciferases/metabolism , Malate Dehydrogenase/metabolism , Models, Biological , Molecular Chaperones/metabolism , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Surface Plasmon Resonance
17.
J Biol Chem ; 281(11): 7479-88, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16415343

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , Escherichia coli/metabolism , Molecular Chaperones/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Anisotropy , Binding Sites , Calorimetry, Differential Scanning , Hot Temperature , Ions , Kinetics , Luciferases/metabolism , Mass Spectrometry , Microscopy, Fluorescence , Molecular Conformation , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity , Temperature , Thermodynamics , Time Factors , Trypsin/chemistry , Trypsin/pharmacology
18.
Protein Sci ; 15(2): 223-33, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16384998

ABSTRACT

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).


Subject(s)
Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Escherichia coli , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Deletion , Spectrophotometry, Infrared , Substrate Specificity
19.
FEBS J ; 272(12): 3184-96, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15955075

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , HSP70 Heat-Shock Proteins/chemistry , Mitochondria/metabolism , Molecular Chaperones/chemistry , Yeasts/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Complementation Test , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Conformation , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Spectrophotometry, Infrared , Yeasts/genetics
20.
J Biol Chem ; 279(19): 19600-6, 2004 May 07.
Article in English | MEDLINE | ID: mdl-14985342

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Binding Sites , Calorimetry, Differential Scanning , Gene Deletion , Infrared Rays , Kinetics , Models, Molecular , Mutation , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Spectrophotometry , Spectrophotometry, Infrared , Temperature , Thermodynamics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...