Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
PNAS Nexus ; 2(4): pgad107, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37091541

ABSTRACT

The ability to assign cellular origin to low-abundance secreted factors in extracellular vesicles (EVs) would greatly facilitate the analysis of paracrine-mediated signaling. Here, we report a method, named selective isolation of extracellular vesicles (SIEVE), which uses cell type-specific proteome labeling via stochastic orthogonal recoding of translation (SORT) to install bioorthogonal reactive groups into the proteins derived from the cells targeted for labeling. We establish the native purification of intact EVs from a target cell, via a bioorthogonal tetrazine ligation, leading to copurification of the largely unlabeled EV proteome from the same cell. SIEVE enables capture of EV proteins at levels comparable with those obtained by antibody-based methods, which capture all EVs regardless of cellular origin, and at levels 20× higher than direct capture of SORT-labeled proteins. Using proteomic analysis, we analyze nonlabeled cargo proteins of EVs and show that the enhanced sensitivity of SIEVE allows for unbiased and comprehensive analysis of EV proteins from subpopulations of cells as well as for cell-specific EV proteomics in complex coculture systems. SIEVE can be applied with high efficiency in a diverse range of existing model systems for cell-cell communication and has direct applications for cell-of-origin EV analysis and for protein biomarker discovery.

2.
Cell Death Dis ; 14(4): 274, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072394

ABSTRACT

The transcription factor p63 shares a high sequence identity with the tumour suppressor p53 which manifests itself in high structural similarity and preference for DNA sequences. Mutations in the DNA binding domain (DBD) of p53 have been studied in great detail, enabling a general mechanism-based classification. In this study we provide a detailed investigation of all currently known mutations in the p63 DBD, which are associated with developmental syndromes, by measuring their impact on transcriptional activity, DNA binding affinity, zinc binding capacity and thermodynamic stability. Some of the mutations we have further characterized with respect to their ability to convert human dermal fibroblasts into induced keratinocytes. Here we propose a classification of the p63 DBD mutations based on the four different mechanisms of DNA binding impairment which we identified: direct DNA contact, zinc finger region, H2 region, and dimer interface mutations. The data also demonstrate that, in contrast to p53 cancer mutations, no p63 mutation induces global unfolding and subsequent aggregation of the domain. The dimer interface mutations that affect the DNA binding affinity by disturbing the interaction between the individual DBDs retain partial DNA binding capacity which correlates with a milder patient phenotype.


Subject(s)
Tumor Suppressor Protein p53 , Tumor Suppressor Proteins , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Protein Binding/genetics , Mutation/genetics , DNA/metabolism , Binding Sites
3.
Mol Oncol ; 15(5): 1277-1288, 2021 05.
Article in English | MEDLINE | ID: mdl-33734563

ABSTRACT

There is a persistent variation in cancer outcomes among and within European countries suggesting (among other causes) inequalities in access to or delivery of high-quality cancer care. European policy (EU Cancer Mission and Europe's Beating Cancer Plan) is currently moving towards a mission-oriented approach addressing these inequalities. In this study, we used the quantitative and qualitative data of the Organisation of European Cancer Institutes' Accreditation and Designation Programme, relating to 40 large European cancer centres, to describe their current compliance with quality standards, to identify the hallmarks common to all centres and to show the distinctive features of Comprehensive Cancer Centres. All Comprehensive Cancer Centres and Cancer Centres accredited by the Organisation of European Cancer Institutes show good compliance with quality standards related to care, multidisciplinarity and patient centredness. However, Comprehensive Cancer Centres on average showed significantly better scores on indicators related to the volume, quality and integration of translational research, such as high-impact publications, clinical trial activity (especially in phase I and phase IIa trials) and filing more patents as early indicators of innovation. However, irrespective of their size, centres show significant variability regarding effective governance when functioning as entities within larger hospitals.


Subject(s)
Cancer Care Facilities , Neoplasms/therapy , Quality of Health Care , Academies and Institutes/standards , Academies and Institutes/statistics & numerical data , Biomedical Research/organization & administration , Biomedical Research/standards , Biomedical Research/statistics & numerical data , Cancer Care Facilities/organization & administration , Cancer Care Facilities/statistics & numerical data , Cohort Studies , Europe/epidemiology , Humans , Medical Oncology/standards , Medical Oncology/statistics & numerical data , Neoplasms/epidemiology , Patient Care Team/organization & administration , Patient Care Team/standards , Patient Care Team/statistics & numerical data , Patient-Centered Care/organization & administration , Patient-Centered Care/standards , Patient-Centered Care/statistics & numerical data , Translational Research, Biomedical/methods , Translational Research, Biomedical/organization & administration , Translational Research, Biomedical/statistics & numerical data
4.
Structure ; 26(8): 1091-1100.e4, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30099987

ABSTRACT

Despite high sequence homology among the p53 family members, the regulation of their transactivation potential is based on strikingly different mechanisms. Previous studies revealed that the activity of TAp63α is regulated via an autoinhibitory mechanism that keeps inactive TAp63α in a dimeric conformation. While all p73 isoforms are constitutive tetramers, their basal activity is much lower compared with tetrameric TAp63. We show that the dimeric state of TAp63α not only reduces DNA binding affinity, but also suppresses interaction with the acetyltransferase p300. Exchange of the transactivation domains is sufficient to transfer the regulatory characteristics between p63 and p73. Structure determination of the transactivation domains of p63 and p73 in complex with the p300 Taz2 domain further revealed that, in contrast to p53 and p73, p63 has a single transactivation domain. Sequences essential for stabilizing the closed dimer of TAp63α have evolved into a second transactivation domain in p73 and p53.


Subject(s)
DNA/chemistry , E1A-Associated p300 Protein/chemistry , Transcription Factors/chemistry , Transcriptional Activation , Tumor Protein p73/chemistry , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Cloning, Molecular , DNA/genetics , DNA/metabolism , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Neurons , Osteoblasts , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Thermodynamics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Nat Struct Mol Biol ; 25(3): 261-269, 2018 03.
Article in English | MEDLINE | ID: mdl-29483652

ABSTRACT

The survival rate of cancer patients is steadily increasing, owing to more efficient therapies. Understanding the molecular mechanisms of chemotherapy-induced premature ovarian insufficiency (POI) could identify targets for prevention of POI. Loss of the primordial follicle reserve is the most important cause of POI, with the p53 family member p63 being responsible for DNA-damage-induced apoptosis of resting oocytes. Here, we provide the first detailed mechanistic insight into the activation of p63, a process that requires phosphorylation by both the priming kinase CHK2 and the executioner kinase CK1 in mouse primordial follicles. We further describe the structural changes induced by phosphorylation that enable p63 to adopt its active tetrameric conformation and demonstrate that previously discussed phosphorylation by c-Abl is not involved in this process. Inhibition of CK1 rescues primary oocytes from doxorubicin and cisplatin-induced apoptosis, thus uncovering a new target for the development of fertoprotective therapies.


Subject(s)
Casein Kinase I/metabolism , Checkpoint Kinase 2/metabolism , DNA Damage , Oocytes/enzymology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Casein Kinase I/antagonists & inhibitors , Cell Line, Tumor , Cisplatin/toxicity , Doxorubicin/toxicity , Humans , Mice , Oocytes/drug effects , Oocytes/metabolism , Phosphorylation , Protein Multimerization
7.
Proc Natl Acad Sci U S A ; 115(5): E906-E915, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29339502

ABSTRACT

The p63 gene encodes a master regulator of epidermal commitment, development, and differentiation. Heterozygous mutations in the C-terminal domain of the p63 gene can cause ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome, a life-threatening disorder characterized by skin fragility and severe, long-lasting skin erosions. Despite deep knowledge of p63 functions, little is known about mechanisms underlying disease pathology and possible treatments. Here, we show that multiple AEC-associated p63 mutations, but not those causative of other diseases, lead to thermodynamic protein destabilization, misfolding, and aggregation, similar to the known p53 gain-of-function mutants found in cancer. AEC mutant proteins exhibit impaired DNA binding and transcriptional activity, leading to dominant negative effects due to coaggregation with wild-type p63 and p73. Importantly, p63 aggregation occurs also in a conditional knock-in mouse model for the disorder, in which the misfolded p63 mutant protein leads to severe epidermal defects. Variants of p63 that abolish aggregation of the mutant proteins are able to rescue p63's transcriptional function in reporter assays as well as in a human fibroblast-to-keratinocyte conversion assay. Our studies reveal that AEC syndrome is a protein aggregation disorder and opens avenues for therapeutic intervention.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Eye Abnormalities/genetics , Phosphoproteins/genetics , Skin/pathology , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Ectoderm/metabolism , Frameshift Mutation , HEK293 Cells , Heterozygote , Humans , Mice , Mutation , Mutation, Missense , Protein Binding , Protein Denaturation , Transcription, Genetic
8.
J Cell Sci ; 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28794013

ABSTRACT

Germ cells are totipotent and, in principle, immortal as they are the source for new germ cells in each generation. This very special role requires tight quality control systems. The p53 protein family constitutes one of the most important quality surveillance systems in cells. Whereas p53 has become famous for its role as the guardian of the genome in its function as the most important somatic tumor suppressor, p63 has been nicknamed 'guardian of the female germ line'. p63 is strongly expressed in resting oocytes and responsible for eliminating those that carry DNA double-strand breaks. The third family member, p73, acts later during oocyte and embryo development by ensuring correct assembly of the spindle assembly checkpoint. In addition to its role in the female germ line, p73 regulates cell-cell contacts between developing sperm cells and supporting somatic cells in the male germ line. Here, we review the involvement of the p53 protein family in the development of germ cells with a focus on quality control in the female germ line and discuss medical implications for cancer patients.

9.
EMBO Rep ; 18(5): 733-744, 2017 05.
Article in English | MEDLINE | ID: mdl-28336776

ABSTRACT

Caspases are key enzymes responsible for mediating apoptotic cell death. Across species, caspase-2 is the most conserved caspase and stands out due to unique features. Apart from cell death, caspase-2 also regulates autophagy, genomic stability and ageing. Caspase-2 requires dimerization for its activation which is primarily accomplished by recruitment to high molecular weight protein complexes in cells. Here, we demonstrate that apoptosis inhibitor 5 (API5/AAC11) is an endogenous and direct inhibitor of caspase-2. API5 protein directly binds to the caspase recruitment domain (CARD) of caspase-2 and impedes dimerization and activation of caspase-2. Interestingly, recombinant API5 directly inhibits full length but not processed caspase-2. Depletion of endogenous API5 leads to an increase in caspase-2 dimerization and activation. Consistently, loss of API5 sensitizes cells to caspase-2-dependent apoptotic cell death. These results establish API5/AAC-11 as a direct inhibitor of caspase-2 and shed further light onto mechanisms driving the activation of this poorly understood caspase.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 2/metabolism , Caspase Inhibitors/metabolism , Cysteine Endopeptidases/metabolism , Nuclear Proteins/metabolism , Apoptosis , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Autophagy , Caspase 2/chemistry , Caspase Activation and Recruitment Domain , Cysteine Endopeptidases/chemistry , Enzyme Activation , HeLa Cells , Humans , Mass Spectrometry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Protein Binding , Protein Multimerization
10.
Article in English | MEDLINE | ID: mdl-27091942

ABSTRACT

The family of the p53 tumor suppressive transcription factors includes p73 and p63 in addition to p53 itself. Given the high degree of amino-acid-sequence homology and structural organization shared by the p53 family members, they display some common features (i.e., induction of cell death, cell-cycle arrest, senescence, and metabolic regulation in response to cellular stress) as well as several distinct properties. Here, we describe the structural evolution of the family members with recent advances on the molecular dynamic studies of p53 itself. A crucial role of the carboxy-terminal domain in regulating the properties of the DNA-binding domain (DBD) supports an induced-fit mechanism, in which the binding of p53 on individual promoters is preferentially regulated by the KOFF over KON.


Subject(s)
Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Binding Sites , Humans , Molecular Dynamics Simulation , Promoter Regions, Genetic , Trans-Activators/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
11.
Cell Death Differ ; 23(12): 1952-1960, 2016 12.
Article in English | MEDLINE | ID: mdl-27447112

ABSTRACT

The high percentage of p53 missense mutations found in cancer has been attributed to mutant acquired oncogenic gain of functions. Different aspects of these tumour-promoting functions are caused by repression of the transcriptional activity of p53 family members p63 and p73. A subset of frequently occurring p53 mutations results in thermodynamic destabilisation of the DNA-binding domain (DBD) rendering this domain highly unstable. These conformational mutants (such as p53R175H) have been suggested to directly bind to p63 and p73 via a co-aggregation mechanism mediated by their DBDs. Although the DBDs of p63 and p73 are in fact not sufficient for the interaction as shown previously, we demonstrate here that the transactivation inhibitory (TI) domains within the α-isoform-specific C termini of p63 and p73 are essential for binding to p53R175H. Hence, the closed dimeric conformation of inactive TAp63α that renders the TI domain inaccessible prevents efficient interaction. We further show that binding to p53R175H correlates with an intrinsic aggregation propensity of the tetrameric α-isoforms conferred by an openly accessible TI domain again supporting interaction via a co-aggregation mechanism.


Subject(s)
Mutant Proteins/metabolism , Protein Aggregates , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Protein p73/chemistry , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Humans , Models, Biological , Mutant Proteins/chemistry , Peptides/metabolism , Protein Binding , Protein Domains , Protein Multimerization
12.
Elife ; 52016 03 14.
Article in English | MEDLINE | ID: mdl-27021569

ABSTRACT

Mammalian oocytes are arrested in the dictyate stage of meiotic prophase I for long periods of time, during which the high concentration of the p53 family member TAp63α sensitizes them to DNA damage-induced apoptosis. TAp63α is kept in an inactive and exclusively dimeric state but undergoes rapid phosphorylation-induced tetramerization and concomitant activation upon detection of DNA damage. Here we show that the TAp63α dimer is a kinetically trapped state. Activation follows a spring-loaded mechanism not requiring further translation of other cellular factors in oocytes and is associated with unfolding of the inhibitory structure that blocks the tetramerization interface. Using a combination of biophysical methods as well as cell and ovary culture experiments we explain how TAp63α is kept inactive in the absence of DNA damage but causes rapid oocyte elimination in response to a few DNA double strand breaks thereby acting as the key quality control factor in maternal reproduction.


Subject(s)
DNA Damage , Oocytes/physiology , Phosphoproteins/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Trans-Activators/metabolism , Animals , Apoptosis , Female , Mice , Phosphorylation , Quality Control
13.
Proc Natl Acad Sci U S A ; 109(38): 15312-7, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22949650

ABSTRACT

p63 inhibits metastasis. Here, we show that p63 (both TAp63 and ΔNp63 isoforms) regulates expression of miR-205 in prostate cancer (PCa) cells, and miR-205 is essential for the inhibitory effects of p63 on markers of epithelial-mesenchymal transition (EMT), such as ZEB1 and vimentin. Correspondingly, the inhibitory effect of p63 on EMT markers and cell migration is reverted by anti-miR-205. p53 mutants inhibit expression of both p63 and miR-205, and the cell migration, in a cell line expressing endogenous mutated p53, can be abrogated by pre-miR-205 or silencing of mutated p53. In accordance with this in vitro data, ΔNp63 or miR-205 significantly inhibits the incidence of lung metastasis in vivo in a mouse tail vein model. Similarly, one or both components of the p63/miR-205 axis were absent in metastases or colonized lymph nodes in a set of 218 human prostate cancer samples. This was confirmed in an independent clinical data set of 281 patients. Loss of this axis was associated with higher Gleason scores, an increased likelihood of metastatic and infiltration events, and worse prognosis. These data suggest that p63/miR-205 may be a useful clinical predictor of metastatic behavior in prostate cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Phosphoproteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred BALB C , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , Protein Isoforms
SELECTION OF CITATIONS
SEARCH DETAIL
...