Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Adv Exp Med Biol ; 1270: 31-44, 2021.
Article in English | MEDLINE | ID: mdl-33123991

ABSTRACT

Recently, it has become clearer that tumor plasticity increases the chance that cancer cells could acquire new mechanisms to escape immune surveillance, become resistant to conventional drugs, and spread to distant sites.Effectively, tumor plasticity drives adaptive response of cancer cells to hypoxia and nutrient deprivation leading to stimulation of neoangionesis or tumor escape. Therefore, tumor plasticity is believed to be a great contributor in recurrence and metastatic dissemination of cancer cells. Importantly, it could be an Achilles' heel of cancer if we could identify molecular mechanisms dictating this phenotype.The reactivation of stem-like signalling pathways is considered a great determinant of tumor plasticity; in addition, a key role has been also attributed to tumor microenvironment (TME). Indeed, it has been proved that cancer cells interact with different cells in the surrounding extracellular matrix (ECM). Interestingly, well-established communication represents a potential allied in maintenance of a plastic phenotype in cancer cells supporting tumor growth and spread. An important signalling pathway mediating cancer cell-TME crosstalk is represented by the HGF/c-Met signalling.Here, we review the role of the HGF/c-Met signalling in tumor-stroma crosstalk focusing on novel findings underlying its role in tumor plasticity, immune escape, and development of adaptive mechanisms.


Subject(s)
Hepatocyte Growth Factor/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Tumor Microenvironment , Humans
2.
Aging Clin Exp Res ; 33(4): 747-758, 2021 Apr.
Article in English | MEDLINE | ID: mdl-31583531

ABSTRACT

Alzheimer's disease (AD) is the most frequent neurodegenerative disorder in the elderly, occurring in approximately 20% of people older than 80. The molecular causes of AD are still poorly understood. However, recent studies have shown that Alternative Splicing (AS) is involved in the gene expression reprogramming associated with the functional changes observed in AD patients. In particular, mutations in cis-acting regulatory sequences as well as alterations in the activity and sub-cellular localization of trans-acting splicing factors and components of the spliceosome machinery are associated with splicing abnormalities in AD tissues, which may influence the onset and progression of the disease. In this review, we discuss the current molecular understanding of how alterations in the AS process contribute to AD pathogenesis. Finally, recent therapeutic approaches targeting aberrant AS regulation in AD are also reviewed.


Subject(s)
Alternative Splicing , Alzheimer Disease , Aged , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Mutation , RNA Splicing/genetics , Spliceosomes/genetics , Spliceosomes/metabolism
3.
Cells ; 9(1)2019 Dec 21.
Article in English | MEDLINE | ID: mdl-31877720

ABSTRACT

During tumor progression, hypoxia, nutrient deprivation or changes in the extracellular environment (i.e., induced by anti-cancer drugs) elicit adaptive responses in cancer cells. Cellular plasticity increases the chance that tumor cells may survive in a challenging microenvironment, acquire new mechanisms of resistance to conventional drugs, and spread to distant sites. Re-activation of stem pathways appears as a significant cause of cellular plasticity because it promotes the acquisition of stem-like properties through a profound phenotypic reprogramming of cancer cells. In addition, it is a major contributor to tumor heterogeneity, depending on the coexistence of phenotypically distinct subpopulations in the same tumor bulk. Several cellular mechanisms may drive this fundamental change, in particular, high-throughput sequencing technologies revealed a key role for alternative splicing (AS). Effectively, AS is one of the most important pre-mRNA processes that increases the diversity of transcriptome and proteome in a tissue- and development-dependent manner. Moreover, defective AS has been associated with several human diseases. However, its role in cancer cell plasticity and tumor heterogeneity remains unclear. Therefore, unravelling the intricate relationship between AS and the maintenance of a stem-like phenotype may explain molecular mechanisms underlying cancer cell plasticity and improve cancer diagnosis and treatment.


Subject(s)
Adaptation, Physiological/genetics , Alternative Splicing/physiology , Neoplasms/genetics , Alternative Splicing/genetics , Antineoplastic Agents/metabolism , Epithelial-Mesenchymal Transition/genetics , Humans , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Transcriptome/genetics , Tumor Microenvironment/genetics
4.
Breast Cancer Res ; 20(1): 105, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30180881

ABSTRACT

BACKGROUND: Development of distant metastases involves a complex multistep biological process termed the invasion-metastasis cascade, which includes dissemination of cancer cells from the primary tumor to secondary organs. NOTCH developmental signaling plays a critical role in promoting epithelial-to-mesenchymal transition, tumor stemness, and metastasis. Although all four NOTCH receptors show oncogenic properties, the unique role of each of these receptors in the sequential stepwise events that typify the invasion-metastasis cascade remains elusive. METHODS: We have established metastatic xenografts expressing high endogenous levels of NOTCH3 using estrogen receptor alpha-positive (ERα+) MCF-7 breast cancer cells with constitutive active Raf-1/mitogen-associated protein kinase (MAPK) signaling (vMCF-7Raf-1) and MDA-MB-231 triple-negative breast cancer (TNBC) cells. The critical role of NOTCH3 in inducing an invasive phenotype and poor outcome was corroborated in unique TNBC cells resulting from a patient-derived brain metastasis (TNBC-M25) and in publicly available claudin-low breast tumor specimens collected from participants in the Molecular Taxonomy of Breast Cancer International Consortium database. RESULTS: In this study, we identified an association between NOTCH3 expression and development of metastases in ERα+ and TNBC models. ERα+ breast tumor xenografts with a constitutive active Raf-1/MAPK signaling developed spontaneous lung metastases through the clonal expansion of cancer cells expressing a NOTCH3 reprogramming network. Abrogation of NOTCH3 expression significantly reduced the self-renewal and invasive capacity of ex vivo breast cancer cells, restoring a luminal CD44low/CD24high/ERαhigh phenotype. Forced expression of the mitotic Aurora kinase A (AURKA), which promotes breast cancer metastases, failed to restore the invasive capacity of NOTCH3-null cells, demonstrating that NOTCH3 expression is required for an invasive phenotype. Likewise, pharmacologic inhibition of NOTCH signaling also impaired TNBC cell seeding and metastatic growth. Significantly, the role of aberrant NOTCH3 expression in promoting tumor self-renewal, invasiveness, and poor outcome was corroborated in unique TNBC cells from a patient-derived brain metastasis and in publicly available claudin-low breast tumor specimens. CONCLUSIONS: These findings demonstrate the key role of NOTCH3 oncogenic signaling in the genesis of breast cancer metastasis and provide a compelling preclinical rationale for the design of novel therapeutic strategies that will selectively target NOTCH3 to halt metastatic seeding and to improve the clinical outcomes of patients with breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Receptor, Notch3/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Self Renewal , Female , Humans , MCF-7 Cells , Mice, Nude , Middle Aged , Neoplasm Seeding , RNA Interference , Receptor, Notch3/metabolism , Survival Analysis , Transplantation, Heterologous , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
6.
J Am Med Dir Assoc ; 18(1): 89.e1-89.e8, 2017 01.
Article in English | MEDLINE | ID: mdl-28012505

ABSTRACT

OBJECTIVES: Cognitive frailty, a condition describing the simultaneous presence of physical frailty and mild cognitive impairment, has been recently defined by an international consensus group. We estimated the predictive role of a "reversible" cognitive frailty model on incident dementia, its subtypes, and all-cause mortality in nondemented older individuals. We verified if vascular risk factors or depressive symptoms could modify this predictive role. DESIGN: Longitudinal population-based study with 3.5- and 7-year of median follow-up. SETTING: Eight Italian municipalities included in the Italian Longitudinal Study on Aging. PARTICIPANTS: In 2150 older individuals from the Italian Longitudinal Study on Aging, we operationalized reversible cognitive frailty with the presence of physical frailty and pre-mild cognitive impairment subjective cognitive decline, diagnosed with a self-report measure based on item 14 of the Geriatric Depression Scale. MEASUREMENTS: Incidence of dementia, its subtypes, and all-cause mortality. RESULTS: Over a 3.5-year follow-up, participants with reversible cognitive frailty showed an increased risk of overall dementia [hazard ratio (HR) 2.30, 95% confidence interval (CI) 1.02-5.18], particularly vascular dementia (VaD), and all-cause mortality (HR 1.74, 95% CI 1.07-2.83). Over a 7-year follow-up, participants with reversible cognitive frailty showed an increased risk of overall dementia (HR 2.12, 95% CI 1.12-4.03), particularly VaD, and all-cause mortality (HR 1.39, 95% CI 1.03-2.00). Vascular risk factors and depressive symptoms did not have any effect modifier on the relationship between reversible cognitive frailty and incident dementia and all-cause mortality. CONCLUSIONS: A model of reversible cognitive frailty was a short- and long-term predictor of all-cause mortality and overall dementia, particularly VaD. The absence of vascular risk factors and depressive symptoms did not modify the predictive role of reversible cognitive frailty on these outcomes.


Subject(s)
Aging , Cause of Death , Cognitive Dysfunction , Frailty/psychology , Mortality/trends , Aged , Aged, 80 and over , Dementia , Female , Humans , Italy/epidemiology , Longitudinal Studies , Male , Proportional Hazards Models
7.
PLoS One ; 11(9): e0162407, 2016.
Article in English | MEDLINE | ID: mdl-27632168

ABSTRACT

The integration of data and knowledge from heterogeneous sources can be a key success factor in drug design, drug repurposing and multi-target therapies. In this context, biological networks provide a useful instrument to highlight the relationships and to model the phenomena underlying therapeutic action in cancer. In our work, we applied network-based modeling within a novel bioinformatics pipeline to identify promising multi-target drugs. Given a certain tumor type/subtype, we derive a disease-specific Protein-Protein Interaction (PPI) network by combining different data-bases and knowledge repositories. Next, the application of suitable graph-based algorithms allows selecting a set of potentially interesting combinations of drug targets. A list of drug candidates is then extracted by applying a recent data fusion approach based on matrix tri-factorization. Available knowledge about selected drugs mechanisms of action is finally exploited to identify the most promising candidates for planning in vitro studies. We applied this approach to the case of Triple Negative Breast Cancer (TNBC), a subtype of breast cancer whose biology is poorly understood and that lacks of specific molecular targets. Our "in-silico" findings have been confirmed by a number of in vitro experiments, whose results demonstrated the ability of the method to select candidates for drug repurposing.


Subject(s)
Antineoplastic Agents/therapeutic use , Systems Integration , Triple Negative Breast Neoplasms/drug therapy , Female , Humans , Models, Theoretical , Monte Carlo Method
8.
Nat Cell Biol ; 18(8): 897-909, 2016 08.
Article in English | MEDLINE | ID: mdl-27347849

ABSTRACT

In cancer, the tumour suppressor gene TP53 undergoes frequent missense mutations that endow mutant p53 proteins with oncogenic properties. Until now, a universal mutant p53 gain-of-function program has not been defined. By means of multi-omics: proteome, DNA interactome (chromatin immunoprecipitation followed by sequencing) and transcriptome (RNA sequencing/microarray) analyses, we identified the proteasome machinery as a common target of p53 missense mutants. The mutant p53-proteasome axis globally affects protein homeostasis, inhibiting multiple tumour-suppressive pathways, including the anti-oncogenic KSRP-microRNA pathway. In cancer cells, p53 missense mutants cooperate with Nrf2 (NFE2L2) to activate proteasome gene transcription, resulting in resistance to the proteasome inhibitor carfilzomib. Combining the mutant p53-inactivating agent APR-246 (PRIMA-1MET) with the proteasome inhibitor carfilzomib is effective in overcoming chemoresistance in triple-negative breast cancer cells, creating a therapeutic opportunity for treatment of solid tumours and metastasis with mutant p53.


Subject(s)
Mutant Proteins/drug effects , Mutation, Missense/drug effects , Proteasome Endopeptidase Complex/drug effects , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/pharmacology , Humans , Mice , MicroRNAs/genetics , Mutant Proteins/genetics , Mutation, Missense/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteome/drug effects , Quinuclidines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism
9.
Int J Mol Sci ; 17(1)2016 Jan 16.
Article in English | MEDLINE | ID: mdl-26784191

ABSTRACT

Breast cancer is the most common tumor and the second leading cause of cancer death among woman, mainly caused by the metastatic spread. Tumor invasiveness is due to an altered expression of adhesion molecules. Among them, semaphorins are of peculiar interest. Cancer cells can manipulate alternative splicing patterns to modulate the expression of adhesion- and motility-related molecules, also at the isoform level. In this study, combining RNA-Sequencing on MCF-7 to targeted experimental validations-in human breast cell lines and breast tumor biopsies-we identified 12 new alternative splicing transcripts in genes encoding adhesion- and motility-related molecules, including semaphorins, their receptors and co-receptors. Among them, a new SEMA3F transcript is expressed in all breast cell lines and breast cancer biopsies, and is translated into a new semaphorin 3F isoform. In silico analysis predicted that most of the new putative proteins lack functional domains, potentially missing some functions and acquiring new ones. Our findings better describe the extent of alternative splicing in breast cancer and highlight the need to further investigate adhesion- and motility-related molecules to gain insights into breast cancer progression.


Subject(s)
Alternative Splicing , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Movement , Exons , Female , Humans , Introns , MCF-7 Cells , Membrane Proteins/metabolism , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Alignment , Sequence Analysis, DNA
10.
Mol Cell Proteomics ; 15(1): 109-23, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26527623

ABSTRACT

Cancer is a very heterogeneous disease, and biological variability adds a further level of complexity, thus limiting the ability to identify new genes involved in cancer development. Oncogenes whose expression levels control cell aggressiveness are very useful for developing cellular models that permit differential expression screenings in isogenic contexts. HMGA1 protein has this unique property because it is a master regulator in breast cancer cells that control the transition from a nontumorigenic epithelial-like phenotype toward a highly aggressive mesenchymal-like one. The proteins extracted from HMGA1-silenced and control MDA-MB-231 cells were analyzed using label-free shotgun mass spectrometry. The differentially expressed proteins were cross-referenced with DNA microarray data obtained using the same cellular model and the overlapping genes were filtered for factors linked to poor prognosis in breast cancer gene expression meta-data sets, resulting in an HMGA1 protein signature composed of 21 members (HRS, HMGA1 reduced signature). This signature had a prognostic value (overall survival, relapse-free survival, and distant metastasis-free survival) in breast cancer. qRT-PCR, Western blot, and immunohistochemistry analyses validated the link of three members of this signature (KIFC1, LRRC59, and TRIP13) with HMGA1 expression levels both in vitro and in vivo and wound healing assays demonstrated that these three proteins are involved in modulating tumor cell motility. Combining proteomic and genomic data with the aid of bioinformatic tools, our results highlight the potential involvement in neoplastic transformation of a restricted list of factors with an as-yet-unexplored role in cancer. These factors are druggable targets that could be exploited for the development of new, targeted therapeutic approaches in triple-negative breast cancer.


Subject(s)
Breast Neoplasms/metabolism , HMGA1a Protein/metabolism , Proteome/metabolism , Proteomics/methods , ATPases Associated with Diverse Cellular Activities , Blotting, Western , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Disease-Free Survival , Gene Expression Regulation, Neoplastic , HMGA1a Protein/genetics , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Kinesins/genetics , Kinesins/metabolism , Mass Spectrometry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Multivariate Analysis , Prognosis , Proteome/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Translational Research, Biomedical/methods
11.
Oncotarget ; 5(3): 613-33, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24327602

ABSTRACT

Adipose tissue is a reservoir of Mesenchymal Stem Cells (Adipose-derived Mesenchymal Stem Cells, ASCs), endowed with regenerative properties. Fat graft was proposed for breast reconstruction in post-surgery cancer patients achieving good aesthetic results and tissues regeneration. However, recent findings highlight a potential tumorigenic role that ASCs may have in cancer recurrence, raising some concerns about their safety in clinical application. To address this issue, we established a model where autologous ASCs were combined with primary normal or cancer cells from breast of human donors, in order to evaluate potential effects of their interactions, in vitro and in vivo. Surprisingly, we found that ASCs are not tumorigenic per sè, as they are not able to induce a neoplastic transformation of normal mammary cells, however they could exhacerbate tumorigenic behaviour of c-Met-expressing breast cancer cells, creating an inflammatory microenvironment which sustained tumor growth and angiogenesis. Pharmacological c-Met inhibition showed that a HGF/c-Met crosstalk between ASCs and breast cancer cells enhanced tumor cells migration, acquiring a metastatic signature, and sustained tumor self-renewal. The master role of HGF/c-Met pathway in cancer recurrence was further confirmed by c-Met immunostaining in primary breast cancer from human donors, revealing a strong positivity in patients displaying a recurrent pathology after fat grafts and a weak/moderate staining in patients without signs of recurrence. Altogether our findings, for the first time, suggest c-Met expression, as predictive to evaluate risk of cancer recurrence after autologous fat graft in post-surgery breast cancer patients, increasing the safety of fat graft in clinical application.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/metabolism , Hepatocyte Growth Factor/metabolism , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-met/metabolism , Adipose Tissue/pathology , Animals , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Female , Heterografts , Humans , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Recurrence, Local/metabolism , Signal Transduction , Tumor Cells, Cultured
13.
PLoS One ; 7(5): e36297, 2012.
Article in English | MEDLINE | ID: mdl-22649490

ABSTRACT

Thiazolidinediones (TZDs) act through peroxisome proliferator activated receptor (PPAR) γ to increase insulin sensitivity in type 2 diabetes (T2DM), but deleterious effects of these ligands mean that selective modulators with improved clinical profiles are needed. We obtained a crystal structure of PPARγ ligand binding domain (LBD) and found that the ligand binding pocket (LBP) is occupied by bacterial medium chain fatty acids (MCFAs). We verified that MCFAs (C8-C10) bind the PPARγ LBD in vitro and showed that they are low-potency partial agonists that display assay-specific actions relative to TZDs; they act as very weak partial agonists in transfections with PPARγ LBD, stronger partial agonists with full length PPARγ and exhibit full blockade of PPARγ phosphorylation by cyclin-dependent kinase 5 (cdk5), linked to reversal of adipose tissue insulin resistance. MCFAs that bind PPARγ also antagonize TZD-dependent adipogenesis in vitro. X-ray structure B-factor analysis and molecular dynamics (MD) simulations suggest that MCFAs weakly stabilize C-terminal activation helix (H) 12 relative to TZDs and this effect is highly dependent on chain length. By contrast, MCFAs preferentially stabilize the H2-H3/ß-sheet region and the helix (H) 11-H12 loop relative to TZDs and we propose that MCFA assay-specific actions are linked to their unique binding mode and suggest that it may be possible to identify selective PPARγ modulators with useful clinical profiles among natural products.


Subject(s)
Fatty Acids/metabolism , Models, Molecular , PPAR gamma/chemistry , PPAR gamma/metabolism , Protein Conformation , Thiazolidinediones/metabolism , 3T3 Cells , Animals , Azo Compounds , Crystallization , Fatty Acids/pharmacology , HeLa Cells , Humans , Mice , Molecular Dynamics Simulation , PPAR gamma/agonists , Protein Structure, Tertiary
14.
Ann Allergy Asthma Immunol ; 108(3): 141-4, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22374194

ABSTRACT

BACKGROUND: The constant increase in the elderly population worldwide has led to a greater interest in immunologic responses during aging. Thus, special attention to allergic diseases in elderly people has begun to emerge, but little is known about the effect and features of allergic rhinitis in elderly people. OBJECTIVE: To evaluate the clinical and cytologic characteristics of respiratory allergy and its impact on the quality of life in elderly people. METHODS: Elderly patients with rhinitis referred to our allergy unit during a 3-month period underwent clinical evaluation and responded to the Rhinasthma Questionnaire. All patients also underwent skin prick testing, measurement of total IgE level, and nasal cytologic analysis. The data were compared with a control group of young adults. RESULTS: Fifty-four patients older than 65 years (mean age, 69.3 years) and 89 young adults (mean age, 26.3 years) with allergic rhinitis were studied. The elderly patients had a less positive family history of atopy (P=.02) and had rhinitis plus conjunctivitis more frequently (P=.002) than young adults, whereas the difference between groups in total IgE level was not statistically significant. On nasal cytologic analysis, the differential count of inflammatory cells did not differ between groups, but in the elderly patients the epithelial-goblet cell ratio was decreased. The quality of life in elderly people was more impaired than in young adults (P=.01). CONCLUSION: In elderly people with allergic rhinitis, the clinical characteristics are different and quality of life is more heavily impaired compared with young adults.


Subject(s)
Rhinitis, Allergic, Perennial , Rhinitis, Allergic, Seasonal , Adolescent , Adult , Aged , Aged, 80 and over , Asthma/epidemiology , Asthma/immunology , Female , Humans , Immunoglobulin E/blood , Male , Quality of Life , Rhinitis, Allergic, Perennial/epidemiology , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/pathology , Rhinitis, Allergic, Perennial/physiopathology , Rhinitis, Allergic, Seasonal/epidemiology , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/pathology , Rhinitis, Allergic, Seasonal/physiopathology , Surveys and Questionnaires , Young Adult
15.
Int J Oncol ; 37(5): 1167-76, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20878064

ABSTRACT

Development of chromosomal instability (CIN) and consequent phenotypic heterogeneity represent common events during breast cancer progression. Breast carcinomas harboring extensive chromosomal aberrations display a more aggressive behavior characterized by chemoresistance and the propensity to give rise to distant metastases. The tumor suppressor p53 plays a key role in the maintenance of chromosomal stability and tissue homeostasis through activation of cell cycle checkpoints following DNA damage and control of centrosome duplication that ensures equal chromosome segregation during cell division. Furthermore, p53 suppresses CD44 expression and the acquisition of stem cell-like properties responsible for epithelial to mesenchymal transition (EMT) and metastasis. In this study we employed MCF-7 breast cancer cells with endogenous wild-type p53, an engineered MCF-7 variant (vMCF-7(DNP53)) overexpressing a dominant negative p53val135 mutant, and cells re-cultured from vMCF-7(DNP53) tumor xenografts. We carried out an integrative transcriptome and cytogenetic analysis to characterize the mechanistic linkage between loss of p53 function, EMT and consequent establishment of invasive gene signatures during breast cancer progression. We demonstrate that abrogation of p53 function drives the early transcriptome changes responsible for cell proliferation, EMT and survival, while further transcriptome changes that occur during in vivo tumor progression are mechanistically linked to the development of CIN leading to a more invasive and metastatic breast cancer phenotype. Here we identified distinct novel non-canonical transcriptome networks involved in cell proliferation, EMT, chemoresistance and invasion that arise following abrogation of p53 function in vitro and development of CIN in vivo. These studies also have important translational implications since some of the nodal genes identified here are 'druggable' making them appropriate molecular targets for the treatment of breast carcinomas displaying mutant p53, EMT, CIN and high metastatic potential.


Subject(s)
Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Animals , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Microarray Analysis , Microscopy, Fluorescence , Neoplasm Invasiveness/pathology , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
16.
BJU Int ; 106(6): 772-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20230385

ABSTRACT

OBJECTIVE: To investigate the utility of tumour carbonic anhydrase IX (CAIX) expression and histological features for predicting the outcome in patients with metastatic clear-cell renal cell carcinoma (mRCC) treated with vascular endothelial growth factor (VEGF)-targeted therapy. PATIENTS AND METHODS: We identified 118 patients with mRCC initiating first-line VEGF-targeted therapy, including 94 with clinical and histological data, and available tissue. The primary endpoint was to detect an interaction between sorafenib vs sunitinib treatment and CAIX status on tumour shrinkage. Other treatment outcomes were also assessed. RESULTS: There was heterogeneity in tumour responsiveness to sunitinib or sorafenib according to CAIX status; the mean shrinkage was -17% vs -25% for sunitinib-treated patients with high vs low tumour CAIX expression, compared to -13% vs +9% for sorafenib-treated patients (P interaction, 0.05). A higher tumour clear-cell component was independently associated with greater tumour shrinkage (P= 0.02), response (P= 0.02) and treatment duration (P= 0.02). CONCLUSIONS: Although CAIX expression had no prognostic value in patients with clear-cell mRCC treated with VEGF-targeted therapy, it might be a predictive biomarker for response to sorafenib treatment. Patients with a higher clear-cell component in their tumours are likely to have a superior clinical benefit from VEGF-targeted therapy.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/therapeutic use , Carbonic Anhydrases/metabolism , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Neoplasm Proteins/metabolism , Aged , Benzenesulfonates/therapeutic use , Carbonic Anhydrase IX , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Epidemiologic Methods , Female , Humans , Immunohistochemistry , Indoles/therapeutic use , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Male , Middle Aged , Niacinamide/analogs & derivatives , Phenylurea Compounds , Prognosis , Pyridines/therapeutic use , Pyrroles/therapeutic use , Sorafenib , Sunitinib , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors
17.
Mol Cancer ; 8: 119, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-20003272

ABSTRACT

BACKGROUND: Aneuploidy is a hallmark of most human cancers that arises as a consequence of chromosomal instability and it is frequently associated with centrosome amplification. Functional inactivation of the Retinoblastoma protein (pRb) has been indicated as a cause promoting chromosomal instability as well centrosome amplification. However, the underlying molecular mechanism still remains to be clarified. RESULTS: Here we show that pRb depletion both in wild type and p53 knockout HCT116 cells was associated with the presence of multipolar spindles, anaphase bridges, lagging chromosomes and micronuclei harbouring whole chromosomes. In addition aneuploidy caused by pRb acute loss was not affected by p53 loss.Quantitative real-time RT-PCR showed that pRB depletion altered expression of genes involved in centrosome duplication, kinetochore assembly and in the Spindle Assembly Checkpoint (SAC). However, despite MAD2 up-regulation pRb-depleted cells seemed to have a functional SAC since they arrested in mitosis after treatments with mitotic poisons. Moreover pRb-depleted HCT116 cells showed BRCA1 overexpression that seemed responsible for MAD2 up-regulation.Post-transcriptional silencing of CENPA by RNA interference, resulting in CENP-A protein levels similar to those present in control cells greatly reduced aneuploid cell numbers in pRb-depleted cells. CONCLUSION: Altogether our findings indicate a novel aspect of pRb acute loss that promotes aneuploidy mainly by inducing CENPA overexpression that in turn might induce micronuclei by affecting the correct attachment of spindle microtubules to kinetochores.


Subject(s)
Autoantigens/genetics , Chromosomal Proteins, Non-Histone/genetics , Genomic Instability , Retinoblastoma Protein/genetics , Autoantigens/physiology , Base Sequence , Blotting, Western , Cell Line, Tumor , Centromere Protein A , Chromosomal Proteins, Non-Histone/physiology , DNA Primers , Humans , Microscopy, Fluorescence , RNA Interference , RNA Processing, Post-Transcriptional , Reverse Transcriptase Polymerase Chain Reaction
18.
BMC Cell Biol ; 10: 79, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19883508

ABSTRACT

BACKGROUND: Changes in chromosome number or structure as well as supernumerary centrosomes and multipolar mitoses are commonly observed in human tumors. Thus, centrosome amplification and mitotic checkpoint dysfunctions are believed possible causes of chromosomal instability. The Retinoblastoma tumor suppressor (RB) participates in the regulation of synchrony between DNA synthesis and centrosome duplication and it is involved in transcription regulation of some mitotic genes. Primary human fibroblasts were transfected transiently with short interfering RNA (siRNA) specific for human pRb to investigate the effects of pRb acute loss on chromosomal stability. RESULTS: Acutely pRb-depleted fibroblasts showed altered expression of genes necessary for cell cycle progression, centrosome homeostasis, kinetochore and mitotic checkpoint proteins. Despite altered expression of genes involved in the Spindle Assembly Checkpoint (SAC) the checkpoint seemed to function properly in pRb-depleted fibroblasts. In particular AURORA-A and PLK1 overexpression suggested that these two genes might have a role in the observed genomic instability. However, when they were post-transcriptionally silenced in pRb-depleted fibroblasts we did not observe reduction in the number of aneuploid cells. This finding suggests that overexpression of these two genes did not contribute to genomic instability triggered by RB acute loss although it affected cell proliferation. Acutely pRb-depleted human fibroblasts showed the presence of micronuclei containing whole chromosomes besides the presence of supernumerary centrosomes and aneuploidy. CONCLUSION: Here we show for the first time that RB acute loss triggers centrosome amplification and aneuploidy in human primary fibroblasts. Altogether, our results suggest that pRb-depleted primary human fibroblasts possess an intact spindle checkpoint and that micronuclei, likely caused by mis-attached kinetochores that in turn trigger chromosome segregation errors, are responsible for aneuploidy in primary human fibroblasts where pRb is acutely depleted.


Subject(s)
Aneuploidy , Cell Nucleus/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Cells, Cultured , Centrosome/metabolism , Chromosomal Instability , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mitosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Retinoblastoma Protein/genetics , Polo-Like Kinase 1
19.
PLoS One ; 4(6): e5877, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19517019

ABSTRACT

There is increasing evidence that p63, and specifically DeltaNp63, plays a central role in both development and tumorigenesis by promoting epithelial cell survival. However, few studies have addressed the molecular mechanisms through which such important function is exerted. Fatty acid synthase (FASN), a key enzyme that synthesizes long-chain fatty acids and is involved in both embryogenesis and cancer, has been recently proposed as a direct target of p53 family members, including p63 and p73. Here we show that knockdown of either total or DeltaN-specific p63 isoforms in squamous cell carcinoma (SCC9) or immortalized prostate epithelial (iPrEC) cells caused a decrease in cell viability by inducing apoptosis without affecting the cell cycle. p63 silencing significantly reduced both the expression and the activity of FASN. Importantly, stable overexpression of either FASN or myristoylated AKT (myr-AKT) was able to partially rescue cells from cell death induced by p63 silencing. FASN induced AKT phosphorylation and a significant reduction in cell viability was observed when FASN-overexpressing SCC9 cells were treated with an AKT inhibitor after p63 knockdown, indicating that AKT plays a major role in FASN-mediated survival. Activated AKT did not cause any alteration in the FASN protein levels but induced its activity, suggesting that the rescue from apoptosis documented in the p63-silenced cells expressing myr-AKT cells may be partially mediated by FASN. Finally, we demonstrated that p63 and FASN expression are positively associated in clinical squamous cell carcinoma samples as well as in the developing prostate. Taken together, our findings demonstrate that FASN is a functionally relevant target of p63 and is required for mediating its pro-survival effects.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Fatty Acid Synthases/metabolism , Membrane Proteins/physiology , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Survival , DNA-Binding Proteins/metabolism , Epithelial Cells , Gene Silencing , Humans , Membrane Proteins/metabolism , Models, Biological , Myristic Acids/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Isoforms , Tumor Protein p73 , Tumor Suppressor Proteins/metabolism
20.
Mol Cancer Ther ; 8(3): 626-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19276158

ABSTRACT

The genetic basis for the hereditary leiomyomatosis and renal cell cancer syndrome is germ-line inactivating mutation in the gene for the Krebs/tricarboxylic acid cycle enzyme, fumarate hydratase (FH), the enzyme that converts fumarate to malate. These individuals are predisposed to development of leiomyomas of the skin and uterus as well as highly aggressive kidney cancers. Inhibition of FH should result in significant decrease in oxidative phosphorylation necessitating that glycolysis followed by fermentation of pyruvate to lactate will be required to provide adequate ATP as well as to regenerate NAD+. Moreover, FH deficiency is known to up-regulate expression of hypoxia-inducible factor (HIF)-1alpha by enhancing the stability of HIF transcript. This leads to activation of various HIF-regulated genes including vascular endothelial growth factor and glucose transporter GLUT1 and increased expression of several glycolytic enzymes. Because lactate dehydrogenase-A (LDH-A), also a HIF-1alpha target, promotes fermentative glycolysis (conversion of pyruvate to lactate), a step essential for regenerating NAD+, we asked whether FH-deficient cells would be exquisitely sensitive to LDH-A blockade. Here, we report that hereditary leiomyomatosis and renal cell cancer tumors indeed overexpress LDH-A, that LDH-A inhibition results in increased apoptosis in a cell with FH deficiency and that this effect is reactive oxygen species mediated, and that LDH-A knockdown in the background of FH knockdown results in significant reduction in tumor growth in a xenograft mouse model.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Enzyme Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , L-Lactate Dehydrogenase/antagonists & inhibitors , Leiomyomatosis/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Enzyme Inhibitors/pharmacology , Female , Fumarate Hydratase/genetics , Gene Knockdown Techniques , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney Neoplasms/complications , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Leiomyomatosis/complications , Leiomyomatosis/genetics , Male , Mice , Mice, Nude , Models, Biological , Syndrome , Tumor Cells, Cultured , Validation Studies as Topic , Xenograft Model Antitumor Assays , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...