Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
2.
Nat Commun ; 14(1): 6777, 2023 10 25.
Article En | MEDLINE | ID: mdl-37880212

Reprogramming of amino acid metabolism, sustained by oncogenic signaling, is crucial for cancer cell survival under nutrient limitation. Here we discovered that missense mutant p53 oncoproteins stimulate de novo serine/glycine synthesis and essential amino acids intake, promoting breast cancer growth. Mechanistically, mutant p53, unlike the wild-type counterpart, induces the expression of serine-synthesis-pathway enzymes and L-type amino acid transporter 1 (LAT1)/CD98 heavy chain heterodimer. This effect is exacerbated by amino acid shortage, representing a mutant p53-dependent metabolic adaptive response. When cells suffer amino acids scarcity, mutant p53 protein is stabilized and induces metabolic alterations and an amino acid transcriptional program that sustain cancer cell proliferation. In patient-derived tumor organoids, pharmacological targeting of either serine-synthesis-pathway and LAT1-mediated transport synergizes with amino acid shortage in blunting mutant p53-dependent growth. These findings reveal vulnerabilities potentially exploitable for tackling breast tumors bearing missense TP53 mutations.


Breast Neoplasms , Tumor Suppressor Protein p53 , Female , Humans , Amino Acids/metabolism , Amino Acids, Essential , Breast Neoplasms/pathology , Glycine , Large Neutral Amino Acid-Transporter 1/genetics , Serine , Tumor Suppressor Protein p53/genetics
3.
Cell Rep ; 36(11): 109694, 2021 09 14.
Article En | MEDLINE | ID: mdl-34525372

Chromatin organization plays a crucial role in tissue homeostasis. Heterochromatin relaxation and consequent unscheduled mobilization of transposable elements (TEs) are emerging as key contributors of aging and aging-related pathologies, including Alzheimer's disease (AD) and cancer. However, the mechanisms governing heterochromatin maintenance or its relaxation in pathological conditions remain poorly understood. Here we show that PIN1, the only phosphorylation-specific cis/trans prolyl isomerase, whose loss is associated with premature aging and AD, is essential to preserve heterochromatin. We demonstrate that this PIN1 function is conserved from Drosophila to humans and prevents TE mobilization-dependent neurodegeneration and cognitive defects. Mechanistically, PIN1 maintains nuclear type-B Lamin structure and anchoring function for heterochromatin protein 1α (HP1α). This mechanism prevents nuclear envelope alterations and heterochromatin relaxation under mechanical stress, which is a key contributor to aging-related pathologies.


Drosophila Proteins/metabolism , Heterochromatin/metabolism , Lamin Type B/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Peptidylprolyl Isomerase/metabolism , Stress, Mechanical , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cells, Cultured , Chromobox Protein Homolog 5/genetics , Chromobox Protein Homolog 5/metabolism , DNA Transposable Elements/genetics , Drosophila/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Humans , Lamin Type B/chemistry , Mice , Mice, Inbred C57BL , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Neocortex/cytology , Neocortex/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Envelope/chemistry , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism
4.
Cell Stress ; 4(11): 261-264, 2020 Oct 05.
Article En | MEDLINE | ID: mdl-33150301

TP53 missense mutations are frequent driver events during tumorigenesis. The majority of TP53 mutations are missense and occur within the DNA binding domain of p53, leading to expression of mutant p53 (mut-p53) proteins that not only lose the tumor suppressive functions of the wild-type (wt-p53) form, but can also acquire novel oncogenic features fostering tumor growth, metastasis and chemoresistance. Mut-p53 affects fundamental cellular pathways and functions through different mechanisms, a major one being the alteration of gene expression. In our recent work (Capaci et al., 2020, Nat Commun) we found that mut-p53, via miR-30d, modifies structure and function of the Golgi apparatus (GA) and induces increased rate of trafficking. This culminates in the release of a pro-malignant secretome, which is capable of remodeling the tumor microenvironment (TME), to increase stiffness of the extracellular matrix (ECM), favouring metastatic colonization, as shown by cell-based assays and experiments of metastatic niche preconditioning in mouse xenograft models. This study provides new insights into the mechanisms by which mut-p53, through induction of non-coding RNAs, can exert pro-tumorigenic functions in a non-cell-autonomous fashion, and highlights potential non-invasive biomarkers and therapeutic targets to treat tumors harboring mut-p53 (Figure 1).

5.
Nat Commun ; 11(1): 3945, 2020 08 07.
Article En | MEDLINE | ID: mdl-32770028

TP53 missense mutations leading to the expression of mutant p53 oncoproteins are frequent driver events during tumorigenesis. p53 mutants promote tumor growth, metastasis and chemoresistance by affecting fundamental cellular pathways and functions. Here, we demonstrate that p53 mutants modify structure and function of the Golgi apparatus, culminating in the increased release of a pro-malignant secretome by tumor cells and primary fibroblasts from patients with Li-Fraumeni cancer predisposition syndrome. Mechanistically, interacting with the hypoxia responsive factor HIF1α, mutant p53 induces the expression of miR-30d, which in turn causes tubulo-vesiculation of the Golgi apparatus, leading to enhanced vesicular trafficking and secretion. The mut-p53/HIF1α/miR-30d axis potentiates the release of soluble factors and the deposition and remodeling of the ECM, affecting mechano-signaling and stromal cells activation within the tumor microenvironment, thereby enhancing tumor growth and metastatic colonization.


Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Golgi Apparatus/pathology , Li-Fraumeni Syndrome/genetics , MicroRNAs/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Biopsy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Female , Fibroblasts , Gene Expression Regulation, Neoplastic , Golgi Apparatus/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Li-Fraumeni Syndrome/pathology , Mice , Microtubules/metabolism , Microtubules/pathology , Mutation , Primary Cell Culture , Secretory Vesicles/metabolism , Secretory Vesicles/pathology , Signal Transduction/genetics , Skin/cytology , Skin/pathology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
6.
Anticancer Drugs ; 31(4): 423-430, 2020 04.
Article En | MEDLINE | ID: mdl-31929348

Bromodomain-containing protein 7 (BRD7) is a member of the bromodomain-containing protein family. Previous studies suggest that BRD7 is predominantly localized in the nucleus, wherein it functions as a transcriptional regulator. Several lines of evidence imply a tumour suppressor function for BRD7. However, the importance of BRD7 in the pathogenesis of breast cancer is not well understood. We have investigated the expression, CpG island methylation and subcellular localization of BRD7 in breast cancer cell lines and clinical cases and thereby assessed its prognostic significance by correlating with clinical-pathological features and time-dependent clinical outcomes. We show that nuclear exclusion of BRD7 occurs commonly in breast cancer and is strongly associated with cases expressing wild-type p53. Moreover, clinical outcomes are significantly less favourable in cases with nuclear exclusion or loss of expression than those in which there is nuclear expression of BRD7. Methylation of the CpG island of BRD7 increases in breast cancer relative to normal breast tissue, but there is not an obvious correlation between methylation and reduced expression or between methylation and clinical outcomes. Overall, our results suggest that nuclear exclusion, rather than transcriptional silencing, is a common mechanism by which the tumour suppressor function of wild-type p53 is inhibited in breast cancer, and show that BRD7 is a promising candidate biomarker in breast cancer.


Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Subcellular Fractions/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromosomal Proteins, Non-Histone/genetics , CpG Islands , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Promoter Regions, Genetic , Retrospective Studies , Survival Rate , Tumor Cells, Cultured
7.
Front Oncol ; 10: 614230, 2020.
Article En | MEDLINE | ID: mdl-33505920

TP53 mutations are widespread in human cancers. An expanding body of evidence highlights that, in addition to their manifold cell-intrinsic activities boosting tumor progression, missense p53 mutants enhance the ability of tumor cells to communicate amongst themselves and with the tumor stroma, by affecting both the quality and the quantity of the cancer secretome. In this review, we summarize recent literature demonstrating that mutant p53 enhances the production of growth and angiogenic factors, inflammatory cytokines and chemokines, modulates biochemical and biomechanical properties of the extracellular matrix, reprograms the cell trafficking machinery to enhance secretion and promote recycling of membrane proteins, and affects exosome composition. All these activities contribute to the release of a promalignant secretome with both local and systemic effects, that is key to the ability of mutant p53 to fuel tumor growth and enable metastatic competence. A precise knowledge of the molecular mechanisms underlying the interplay between mutant p53 and the microenvironment is expected to unveil non-invasive biomarkers and actionable targets to blunt tumor aggressiveness.

8.
Cell Death Differ ; 26(2): 199-212, 2019 01.
Article En | MEDLINE | ID: mdl-30538286

Forty years of research have established that the p53 tumor suppressor provides a major barrier to neoplastic transformation and tumor progression by its unique ability to act as an extremely sensitive collector of stress inputs, and to coordinate a complex framework of diverse effector pathways and processes that protect cellular homeostasis and genome stability. Missense mutations in the TP53 gene are extremely widespread in human cancers and give rise to mutant p53 proteins that lose tumor suppressive activities, and some of which exert trans-dominant repression over the wild-type counterpart. Cancer cells acquire selective advantages by retaining mutant forms of the protein, which radically subvert the nature of the p53 pathway by promoting invasion, metastasis and chemoresistance. In this review, we consider available evidence suggesting that mutant p53 proteins can favor cancer cell survival and tumor progression by acting as homeostatic factors that sense and protect cancer cells from transformation-related stress stimuli, including DNA lesions, oxidative and proteotoxic stress, metabolic inbalance, interaction with the tumor microenvironment, and the immune system. These activities of mutant p53 may explain cancer cell addiction to this particular oncogene, and their study may disclose tumor vulnerabilities and synthetic lethalities that could be exploited for hitting tumors bearing missense TP53 mutations.


Cell Transformation, Neoplastic/genetics , Mutant Proteins/metabolism , Neoplasms/metabolism , Oncogenes , Tumor Suppressor Protein p53/genetics , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Mutation, Missense , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Microenvironment/genetics
10.
Nat Cell Biol ; 20(1): 28-35, 2018 01.
Article En | MEDLINE | ID: mdl-29255172

Tumour-associated p53 missense mutants act as driver oncogenes affecting cancer progression, metastatic potential and drug resistance (gain-of-function) 1 . Mutant p53 protein stabilization is a prerequisite for gain-of-function manifestation; however, it does not represent an intrinsic property of p53 mutants, but rather requires secondary events 2 . Moreover, mutant p53 protein levels are often heterogeneous even within the same tumour, raising questions on the mechanisms that control local mutant p53 accumulation in some tumour cells but not in their neighbours 2,3 . By investigating the cellular pathways that induce protection of mutant p53 from ubiquitin-mediated proteolysis, we found that HDAC6/Hsp90-dependent mutant p53 accumulation is sustained by RhoA geranylgeranylation downstream of the mevalonate pathway, as well as by RhoA- and actin-dependent transduction of mechanical inputs, such as the stiffness of the extracellular environment. Our results provide evidence for an unpredicted layer of mutant p53 regulation that relies on metabolic and mechanical cues.


Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mechanotransduction, Cellular/genetics , Mevalonic Acid/metabolism , Tumor Suppressor Protein p53/genetics , rhoA GTP-Binding Protein/genetics , Actins/genetics , Actins/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Databases, Factual , Female , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6/genetics , Histone Deacetylase 6/metabolism , Humans , Mice , Mice, SCID , Mutation , Protein Stability , Proteolysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/metabolism
11.
FEBS J ; 284(6): 837-850, 2017 03.
Article En | MEDLINE | ID: mdl-27808469

The TP53 tumor suppressor is the most frequently mutated gene in human cancers. In recent years, a blooming of research efforts based on both cell lines and mouse models have highlighted how deeply mutant p53 proteins affect fundamental cellular pathways with cancer-promoting outcomes. Neomorphic mutant p53 activities spread over multiple levels, impinging on chromatin structure, transcriptional regulation and microRNA maturation, shaping the proteome and the cell's metabolic pathways, and also exerting cytoplasmic functions and displaying cell-extrinsic effects. These tumorigenic activities are inextricably linked with the blend of highly corrupted processes that characterize the tumor context. Recent studies indicate that successful strategies to extract core aspects of mutant p53 oncogenic potential and to identify unique tumor dependencies entail the superimposition of large-scale analyses performed in multiple experimental systems, together with a mindful use of animal models. This will hopefully soon lead to the long-awaited inclusion of mutant p53 as an actionable target of clinical antitumor therapies.


Molecular Targeted Therapy , Mutant Proteins/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , MicroRNAs/genetics , Mutant Proteins/therapeutic use , Neoplasms/therapy , Precision Medicine , Signal Transduction , Tumor Suppressor Protein p53/therapeutic use
12.
Front Cell Neurosci ; 10: 110, 2016.
Article En | MEDLINE | ID: mdl-27199664

Huntington's disease (HD) is a fatal, dominantly inherited, neurodegenerative disorder due to a pathological expansion of the CAG repeat in the coding region of the HTT gene. In the quest for understanding the molecular basis of neurodegeneration, we have previously demonstrated that the prolyl isomerase Pin1 plays a crucial role in mediating p53-dependent apoptosis triggered by mutant huntingtin (mHtt) in vitro. To assess the effects of the lack of Pin1 in vivo, we have bred Pin1 knock-out mice with Hdh(Q111) knock-in mice, a genetically precise model of HD. We show that Pin1 genetic ablation modifies a portion of Hdh(Q111) phenotypes in a time-dependent fashion. As an early event, Pin1 activity reduces the DNA damage response (DDR). In midlife mice, by taking advantage of next-generation sequencing technology, we show that Pin1 activity modulates a portion of the alterations triggered by mHtt, extending the role of Pin1 to two additional Hdh(Q111) phenotypes: the unbalance in the "synthesis/concentration of hormones", as well as the alteration of "Wnt/ß-catenin signaling". In aging animals, Pin1 significantly increases the number of mHtt-positive nuclear inclusions while it reduces gliosis. In summary, this work provides further support for a role of Pin1 in HD pathogenesis.

13.
Biochim Biophys Acta ; 1850(10): 2048-60, 2015 Oct.
Article En | MEDLINE | ID: mdl-25641576

BACKGROUND: The p53 protein family, comprising p53, p63 and p73, is primarily involved in preserving genome integrity and preventing tumor onset, and also affects a range of physiological processes. Signal-dependent modifications of its members and of other pathway components provide cells with a sophisticated code to transduce a variety of stress signaling into appropriate responses. TP53 mutations are highly frequent in cancer and lead to the expression of mutant p53 proteins that are endowed with oncogenic activities and sensitive to stress signaling. SCOPE OF REVIEW: p53 family proteins have unique structural and functional plasticity, and here we discuss the relevance of prolyl-isomerization to actively shape these features. MAJOR CONCLUSIONS: The anti-proliferative functions of the p53 family are carefully activated upon severe stress and this involves the interaction with prolyl-isomerases. In particular, stress-induced stabilization of p53, activation of its transcriptional control over arrest- and cell death-related target genes and of its mitochondrial apoptotic function, as well as certain p63 and p73 functions, all require phosphorylation of specific S/T-P motifs and their subsequent isomerization by the prolyl-isomerase Pin1. While these functions of p53 counteract tumorigenesis, under some circumstances their activation by prolyl-isomerases may have negative repercussions (e.g. tissue damage induced by anticancer therapies and ischemia-reperfusion, neurodegeneration). Moreover, elevated Pin1 levels in tumor cells may transduce deregulated phosphorylation signaling into activation of mutant p53 oncogenic functions. GENERAL SIGNIFICANCE: The complex repertoire of biological outcomes induced by p53 finds mechanistic explanations, at least in part, in the association between prolyl-isomerases and the p53 pathway. This article is part of a Special Issue entitled Proline-directed foldases: Cell signaling catalysts and drug targets.


Apoptosis , Cell Proliferation , Peptidylprolyl Isomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Motifs , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Stability , Protein Structure, Tertiary , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Mitochondrion ; 19 Pt A: 88-96, 2014 Nov.
Article En | MEDLINE | ID: mdl-25132079

Mitochondria are sensitive and efficient organelles that regulate essential biological processes including: energy metabolism, decoding and transduction of intracellular signals, and balance between cell death and survival. Of note, dysfunctions in mitochondrial physiology are a general hallmark of cancer cells, leading to transformation-related features such as altered cellular metabolism, survival under stress conditions and reduced apoptotic response to chemotherapy. Mitochondrial apoptosis is a finely regulated process that derives from activation of multiple signaling networks. A crucial biochemical requirement for transducing pro-apoptotic stimuli is represented by kinase-dependent phosphorylation cascades. In this context a pivotal role is played by the prolyl-isomerase Pin1, which translates Ser/Thr-Pro phosphorylation into conformational changes able to modify the activities of its substrates. In this review we will discuss the impact of Pin1 in regulating various aspects of apoptosis in different biological contexts with particular emphasis on cancer and neurodegenerative diseases.


Apoptosis/physiology , Mitochondria/physiology , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Peptidylprolyl Isomerase/metabolism , Gene Expression Regulation/physiology , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics
15.
Proc Natl Acad Sci U S A ; 110(45): E4203-12, 2013 Nov 05.
Article En | MEDLINE | ID: mdl-24145406

Excessive genome damage activates the apoptosis response. Protein kinase HIPK2 is a key regulator of DNA damage-induced apoptosis. Here, we deciphered the molecular mechanism of HIPK2 activation and show its relevance for DNA damage-induced apoptosis in cellulo and in vivo. HIPK2 autointeracts and site-specifically autophosphorylates upon DNA damage at Thr880/Ser882. Autophosphorylation regulates HIPK2 activity and mutation of the phosphorylation-acceptor sites deregulates p53 Ser46 phosphorylation and apoptosis in cellulo. Moreover, HIPK2 autophosphorylation is conserved between human and zebrafish and is important for DNA damage-induced apoptosis in vivo. Mechanistically, autophosphorylation creates a binding signal for the phospho-specific isomerase Pin1. Pin1 links HIPK2 activation to its stabilization by inhibiting HIPK2 polyubiquitination and modulating Siah-1-HIPK2 interaction. Concordantly, Pin1 is required for DNA damage-induced HIPK2 stabilization and p53 Ser46 phosphorylation and is essential for induction of apotosis both in cellulo and in zebrafish. Our results identify an evolutionary conserved mechanism regulating DNA damage-induced apoptosis.


Apoptosis/physiology , Carrier Proteins/metabolism , DNA Damage/physiology , Enzyme Activation/physiology , Peptidylprolyl Isomerase/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Line , Genetic Vectors , Humans , Microscopy, Fluorescence , NIMA-Interacting Peptidylprolyl Isomerase , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics
16.
Proc Natl Acad Sci U S A ; 108(44): 17979-84, 2011 Nov 01.
Article En | MEDLINE | ID: mdl-22011578

Huntington disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the gene coding for huntingtin protein. Several mechanisms have been proposed by which mutant huntingtin (mHtt) may trigger striatal neurodegeneration, including mitochondrial dysfunction, oxidative stress, and apoptosis. Furthermore, mHtt induces DNA damage and activates a stress response. In this context, p53 plays a crucial role in mediating mHtt toxic effects. Here we have dissected the pathway of p53 activation by mHtt in human neuronal cells and in HD mice, with the aim of highlighting critical nodes that may be pharmacologically manipulated for therapeutic intervention. We demonstrate that expression of mHtt causes increased phosphorylation of p53 on Ser46, leading to its interaction with phosphorylation-dependent prolyl isomerase Pin1 and consequent dissociation from the apoptosis inhibitor iASPP, thereby inducing the expression of apoptotic target genes. Inhibition of Ser46 phosphorylation by targeting homeodomain-interacting protein kinase 2 (HIPK2), PKCδ, or ataxia telangiectasia mutated kinase, as well as inhibition of the prolyl isomerase Pin1, prevents mHtt-dependent apoptosis of neuronal cells. These results provide a rationale for the use of small-molecule inhibitors of stress-responsive protein kinases and Pin1 as a potential therapeutic strategy for HD treatment.


Apoptosis/physiology , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Serine/metabolism , Tumor Suppressor Protein p53/physiology , Animals , DNA Damage , Humans , Huntingtin Protein , Isomerism , Mice , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oxidative Stress , Phosphorylation
17.
Cell Cycle ; 9(14): 2777-81, 2010 Jul 15.
Article En | MEDLINE | ID: mdl-20647772

Oncogene-induced senescence (OIS) is a cellular defense mechanism against excessive mitogenic signaling and tumorigenesis. One of the major pathways required for OIS is the p53 tumor suppressor pathway. Consequently, many human tumors harbor p53 mutations while others show a dysfunctional p53 pathway, frequently by unknown mechanisms. We recently identified BRD7 as a potential tumor suppressor gene acting as a transcriptional cofactor for p53, affecting histone acetylation, p53 acetylation, and promoter activity on a subset of p53 target genes. We further found low BRD7 expression specifically in a subgroup of human breast tumors harboring wild-type, but not mutant, p53 and showed that one of the responsible mechanisms is deletion of the BRD7 gene locus. Here we further discuss the role of BRD7 as a cofactor in transcriptional regulation and highlight its role as a tumor suppressor via association with p53 and other tumor suppressor proteins.


Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Tumor Suppressor Proteins/metabolism , Acetylation , Cell Proliferation , Cellular Senescence , Chromosomal Proteins, Non-Histone/chemistry , DNA Repair , Humans , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/chemistry
18.
Nat Cell Biol ; 12(4): 380-9, 2010 Apr.
Article En | MEDLINE | ID: mdl-20228809

Oncogene-induced senescence is a p53-dependent defence mechanism against uncontrolled proliferation. Consequently, many human tumours harbour p53 mutations and others show a dysfunctional p53 pathway, frequently by unknown mechanisms. Here we identify BRD7 (bromodomain-containing 7) as a protein whose inhibition allows full neoplastic transformation in the presence of wild-type p53. In human breast tumours harbouring wild-type, but not mutant, p53 the BRD7 gene locus was frequently deleted and low BRD7 expression was found in a subgroup of tumours. Functionally, BRD7 is required for efficient p53-mediated transcription of a subset of target genes. BRD7 interacts with p53 and p300 and is recruited to target gene promoters, affecting histone acetylation, p53 acetylation and promoter activity. Thus, BRD7 suppresses tumorigenicity by serving as a p53 cofactor required for the efficient induction of p53-dependent oncogene-induced senescence.


Breast Neoplasms/genetics , Chromosomal Proteins, Non-Histone/genetics , Genes, Tumor Suppressor , Tumor Suppressor Protein p53/genetics , Acetylation , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Chromosomal Proteins, Non-Histone/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, ras , Histones/metabolism , Humans , Mutation , Promoter Regions, Genetic , Protein Binding , RNA Interference , Recombinant Fusion Proteins/metabolism , Time Factors , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors/metabolism
19.
Nat Struct Mol Biol ; 14(10): 912-20, 2007 Oct.
Article En | MEDLINE | ID: mdl-17906639

The tumor-suppressor function of p53 relies on its transcriptional activity, which is modulated by post-translational modifications and interactions with regulatory proteins. The prolyl isomerase Pin1 has a central role in transducing phosphorylation of p53 into conformational changes that affect p53 stability and function. We found that Pin1 is required for efficient loading of p53 on target promoters upon stress. In addition, Pin1 is recruited to chromatin by p53 and stimulates binding of the p300 acetyltransferase and consequent p53 acetylation. Accordingly, tumor-associated mutations at Pin1-binding residues within the p53 proline-rich domain hamper acetylation of p53 by p300. After phosphorylation of p53 at Ser46 triggered by cytotoxic stimuli, Pin1 also mediates p53's dissociation from the apoptosis inhibitor iASPP, promoting cell death. In tumors bearing wild-type p53, expression of Pin1 and iASPP are inversely correlated, supporting the clinical relevance of these interactions.


Intracellular Signaling Peptides and Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Processing, Post-Translational , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Acetylation , Apoptosis/physiology , Cell Line, Tumor , Chromatin/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasms/genetics , Neoplasms/metabolism , Peptidylprolyl Isomerase/genetics , Phosphorylation , Polymorphism, Genetic , Promoter Regions, Genetic , Protein Binding , Repressor Proteins , Tumor Suppressor Protein p53/genetics , p300-CBP Transcription Factors/metabolism
20.
Science ; 315(5812): 659-63, 2007 Feb 02.
Article En | MEDLINE | ID: mdl-17272725

The 66-kilodalton isoform of the growth factor adapter Shc (p66Shc) translates oxidative damage into cell death by acting as reactive oxygen species (ROS) producer within mitochondria. However, the signaling link between cellular stress and mitochondrial proapoptotic activity of p66Shc was not known. We demonstrate that protein kinase C beta, activated by oxidative conditions in the cell, induces phosphorylation of p66Shc and triggers mitochondrial accumulation of the protein after it is recognized by the prolyl isomerase Pin1. Once imported, p66Shc causes alterations of mitochondrial Ca2+ responses and three-dimensional structure, thus inducing apoptosis. These data identify a signaling route that activates an apoptotic inducer shortening the life span and could be a potential target of pharmacological approaches to inhibit aging.


Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Cellular Senescence , Mitochondria/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Kinase C/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Calcium Signaling , Cell Survival , Cells, Cultured , Cyclosporine/pharmacology , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Mice , Mitochondria/ultrastructure , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Oxidative Stress , Permeability , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C beta , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
...