Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 36(11): 109694, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525372

ABSTRACT

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.


Subject(s)
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
2.
Cancers (Basel) ; 12(12)2020 Dec 21.
Article in English | MEDLINE | ID: mdl-33371412

ABSTRACT

Tumor organoids are tridimensional cell culture systems that are generated in vitro from surgically resected patients' tumors. They can be propagated in culture maintaining several features of the tumor of origin, including cellular and genetic heterogeneity, thus representing a promising tool for precision cancer medicine. Here, we established patient-derived tumor organoids (PDOs) from different breast cancer subtypes (luminal A, luminal B, human epidermal growth factor receptor 2 (HER2)-enriched, and triple negative). The established model systems showed histological and genomic concordance with parental tumors. However, in PDOs, the ratio of diverse cell populations was frequently different from that originally observed in parental tumors. We showed that tumor organoids represent a valuable system to test the efficacy of standard therapeutic treatments and to identify drug resistant populations within tumors. We also report that inhibitors of mechanosignaling and of Yes-associated protein 1 (YAP) activation can restore chemosensitivity in drug resistant tumor organoids.

3.
Nat Commun ; 11(1): 3945, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770028

ABSTRACT

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.


Subject(s)
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
4.
Front Oncol ; 9: 94, 2019.
Article in English | MEDLINE | ID: mdl-30873382

ABSTRACT

Cellular choices are determined by developmental and environmental stimuli through integrated signal transduction pathways. These critically depend on attainment of proper activation levels that in turn rely on post-translational modifications (PTMs) of single pathway members. Among these PTMs, post-phosphorylation prolyl-isomerization mediated by PIN1 represents a unique mechanism of spatial, temporal and quantitative control of signal transduction. Indeed PIN1 was shown to be crucial for determining activation levels of several pathways and biological outcomes downstream to a plethora of stimuli. Of note, studies performed in different model organisms and humans have shown that hormonal, nutrient, and oncogenic stimuli simultaneously affect both PIN1 activity and the pathways that depend on PIN1-mediated prolyl-isomerization, suggesting the existence of evolutionarily conserved molecular circuitries centered on this isomerase. This review focuses on molecular mechanisms and cellular processes like proliferation, metabolism, and stem cell fate, that are regulated by PIN1 in physiological conditions, discussing how these are subverted in and hijacked by cancer cells. Current status and open questions regarding the use of PIN1 as biomarker and target for cancer therapy as well as clinical development of PIN1 inhibitors are also addressed.

5.
Oncotarget ; 9(29): 20508-20523, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29755668

ABSTRACT

NRF2 (NFE2L2) is one of the main regulators of the antioxidant response of the cell. Here we show that in cancer cells NRF2 targets are selectively upregulated or repressed through a mutant p53-dependent mechanism. Mechanistically, mutant p53 interacts with NRF2, increases its nuclear presence and resides with NRF2 on selected ARE containing gene promoters activating the transcription of a specific set of genes while leading to the transcriptional repression of others. We show that thioredoxin (TXN) is a mutant p53-activated NRF2 target with pro-survival and pro-migratory functions in breast cancer cells under oxidative stress, while heme oxygenase 1 (HMOX1) is a mutant p53-repressed target displaying opposite effects. A gene signature of NRF2 targets activated by mutant p53 shows a significant association with bad overall prognosis and with mutant p53 status in breast cancer patients. Concomitant inhibition of thioredoxin system with Auranofin and of mutant p53 with APR-246 synergizes in killing cancer cells expressing p53 gain-of-function mutants.

6.
Nat Commun ; 8: 15772, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28598431

ABSTRACT

The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Enzyme Inhibitors/chemistry , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Reactive Oxygen Species/metabolism
7.
Cell Death Differ ; 24(2): 200-211, 2017 02.
Article in English | MEDLINE | ID: mdl-27834957

ABSTRACT

Mammary gland development, various stages of mammary tumorigenesis and breast cancer progression have the peptidyl-prolyl cis/trans isomerase PIN1 at their centerpiece, in virtue of the ability of this unique enzyme to fine-tune the dynamic crosstalk between multiple molecular pathways. PIN1 exerts its action by inducing conformational and functional changes on key cellular proteins, following proline-directed phosphorylation. Through this post-phosphorylation signal transduction mechanism, PIN1 controls the extent and direction of the cellular response to a variety of inputs, in physiology and disease. This review discusses PIN1's roles in normal mammary development and cancerous progression, as well as the clinical impact of targeting this enzyme in breast cancer patients.


Subject(s)
Breast Neoplasms/pathology , Mammary Glands, Human/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , E2F Transcription Factors/metabolism , Enzyme Inhibitors/therapeutic use , Female , Genomic Instability , Humans , Mammary Glands, Human/growth & development , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL