28 results on '"Folgiero, Valentina"'
Search Results
2. GMP-manufactured CRISPR/Cas9 technology as an advantageous tool to support cancer immunotherapy
- Author
-
Caforio, M, Iacovelli, S, Quintarelli, C, Locatelli, F, and Folgiero, Valentina
- Published
- 2024
- Full Text
- View/download PDF
3. Manipulating the tumor immune microenvironment to improve cancer immunotherapy: IGF1R, a promising target.
- Author
-
Pellegrino, Marsha, Secli, Valerio, D’Amico, Silvia, Petrilli, Lucia Lisa, Caforio, Matteo, Folgiero, Valentina, Tumino, Nicola, Vacca, Paola, Vinci, Maria, Fruci, Doriana, and de Billy, Emmanuel
- Subjects
TUMOR microenvironment ,INSULIN-like growth factor receptors ,SOMATOMEDIN C ,IMMUNOTHERAPY ,CELL physiology - Abstract
Cancer immunotherapy has made impressive advances in improving the outcome of patients affected by malignant diseases. Nonetheless, some limitations still need to be tackled to more efficiently and safely treat patients, in particular for those affected by solid tumors. One of the limitations is related to the immunosuppressive tumor microenvironment (TME), which impairs antitumor immunity. Efforts to identify targets able to turn the TME into a milieu more auspicious to current immuno-oncotherapy is a real challenge due to the high redundancy of the mechanisms involved. However, the insulin-like growth factor 1 receptor (IGF1R), an attractive drug target for cancer therapy, is emerging as an important immunomodulator and regulator of key immune cell functions. Here, after briefly summarizing the IGF1R signaling pathway in cancer, we review its role in regulating immune cells function and activity, and discuss IGF1R as a promising target to improve anti-cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. AATF/Che-1 RNA polymerase II binding protein overexpression reduces the anti-tumor NK-cell cytotoxicity through activating receptors modulation.
- Author
-
Caforio, Matteo, Tumino, Nicola, Sorino, Cristina, Manni, Isabella, Di Giovenale, Stefano, Piaggio, Giulia, Iezzi, Simona, Strimpakos, Georgios, Mattei, Elisabetta, Moretta, Lorenzo, Fanciulli, M., Vacca, Paola, Locatelli, Franco, and Folgiero, Valentina
- Subjects
RNA polymerase II ,CARRIER proteins ,KILLER cells ,GENETIC overexpression ,CELL receptors - Abstract
Introduction: AATF/Che-1 over-expression in different tumors is well known and its effect on tumorigenicity is mainly due to its central role demonstrated in the oncogenic pathways of solid tumors, where it controls proliferation and viability. The effect exerted by tumors overexpressing Che-1 on the immune response has not yet been investigated. Methods: Starting from ChIP-sequencing data we confirmed Che-1 enrichment on Nectin-1 promoter. Several co-cultures experiments between NK-cells and tumor cells transduced by lentiviral vectors carrying Che-1-interfering sequence, analyzed by flow-cytometry have allowed a detailed characterization of NK receptors and tumor ligands expression. Results: Here, we show that Che-1 is able to modulate the expression of Nectin-1 ligand at the transcriptional level, leading to the impairment of killing activity of NK-cells. Nectin-1 down-modulation induces a modification in NK-cell ligands expression able to interact with activating receptors and to stimulate NK-cell function. In addition, NK-cells from Che-1 transgenic mice, confirming a reduced expression of activating receptors, exhibit impaired activation and a preferential immature status. Discussion: The critical equilibrium between NK-cell ligand expression on tumor cells and the interaction with NK cell receptors is affected by Che-1 over-expression and partially restored by Che-1 interference. The evidence of a new role for Che-1 as regulator of anti-tumor immunity supports the necessity to develop approaches able to target this molecule which shows a dual tumorigenic function as cancer promoter and immune response modulator. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Recent advances in searching c-Myc transcriptional cofactors during tumorigenesis
- Author
-
Caforio, Matteo, Sorino, Cristina, Iacovelli, Stefano, Fanciulli, Maurizio, Locatelli, Franco, and Folgiero, Valentina
- Published
- 2018
- Full Text
- View/download PDF
6. Che‐1 is targeted by c‐Myc to sustain proliferation in pre‐B‐cell acute lymphoblastic leukemia
- Author
-
Folgiero, Valentina, Sorino, Cristina, Pallocca, Matteo, De Nicola, Francesca, Goeman, Frauke, Bertaina, Valentina, Strocchio, Luisa, Romania, Paolo, Pitisci, Angela, Iezzi, Simona, Catena, Valeria, Bruno, Tiziana, Strimpakos, Georgios, Passananti, Claudio, Mattei, Elisabetta, Blandino, Giovanni, Locatelli, Franco, and Fanciulli, Maurizio
- Published
- 2018
- Full Text
- View/download PDF
7. Metastatic Group 3 Medulloblastoma in a Patient With Tuberous Sclerosis Complex: Case Description and Molecular Characterization of the Tumor
- Author
-
Moavero, Romina, Folgiero, Valentina, Carai, Andrea, Miele, Evelina, Ferretti, Elisabetta, Po, Agnese, Camassei, Francesca Diomedi, Lepri, Francesca Romana, Vigevano, Federico, Curatolo, Paolo, Valeriani, Massimiliano, Colafati, Giovanna S., Locatelli, Franco, Tornesello, Assunta, and Mastronuzzi, Angela
- Published
- 2016
- Full Text
- View/download PDF
8. Indoleamine 2,3-dioxygenase 1 (IDO1) activity correlates with immune system abnormalities in multiple myeloma
- Author
-
Bonanno Giuseppina, Mariotti Andrea, Procoli Annabella, Folgiero Valentina, Natale Daniela, De Rosa Luca, Majolino Ignazio, Novarese Linda, Rocci Alberto, Gambella Manuela, Ciciarello Marilena, Scambia Giovanni, Palumbo Antonio, Locatelli Franco, De Cristofaro Raimondo, and Rutella Sergio
- Subjects
Medicine - Abstract
Abstract Background Multiple myeloma (MM) is a plasma cell malignancy with a multifaceted immune dysfunction. Indoleamine 2,3-dioxygenase 1 (IDO1) degrades tryptophan into kynurenine (KYN), which inhibits effector T cells and promote regulatory T-cell (Treg) differentiation. It is presently unknown whether MM cells express IDO1 and whether IDO1 activity correlates with immune system impairment. Methods We investigated IDO1 expression in 25 consecutive patients with symptomatic MM and in 7 patients with either monoclonal gammopathy of unknown significance (MGUS; n=3) or smoldering MM (SMM; n=4). IDO1-driven tryptophan breakdown was correlated with the release of hepatocyte growth factor (HGF) and with the frequency of Treg cells and NY-ESO-1-specific CD8+ T cells. Results KYN was increased in 75% of patients with symptomatic MM and correlated with the expansion of CD4+CD25+FoxP3+ Treg cells and the contraction of NY-ESO-1-specific CD8+ T cells. In vitro, primary MM cells promoted the differentiation of allogeneic CD4+ T cells into bona fide CD4+CD25hiFoxP3hi Treg cells and suppressed IFN-γ/IL-2 secretion, while preserving IL-4 and IL-10 production. Both Treg expansion and inhibition of Th1 differentiation by MM cells were reverted, at least in part, by d,l-1-methyl-tryptophan, a chemical inhibitor of IDO. Notably, HGF levels were higher within the BM microenvironment of patients with IDO+ myeloma disease compared with patients having IDO- MM. Mechanistically, the antagonism of MET receptor for HGF with SU11274, a MET inhibitor, prevented HGF-induced AKT phosphorylation in MM cells and translated into reduced IDO protein levels and functional activity. Conclusions These data suggest that IDO1 expression may contribute to immune suppression in patients with MM and possibly other HGF-producing cancers.
- Published
- 2012
- Full Text
- View/download PDF
9. Upfront treatment with mTOR inhibitor everolimus in pediatric low-grade gliomas: A single-center experience.
- Author
-
Cacchione, Antonella, Lodi, Mariachiara, Carai, Andrea, Miele, Evelina, Tartaglia, Marco, Megaro, Giacomina, Baldo, Giada Del, Alessi, Iside, Colafati, Giovanna Stefania, Carboni, Alessia, Boccuto, Luigi, Camassei, Francesca Diomedi, Catanzaro, Giuseppina, Po, Agnese, Ferretti, Elisabetta, Pedace, Lucia, Pizzi, Simone, Folgiero, Valentina, Pezzullo, Marco, and Corsetti, Tiziana
- Subjects
EVEROLIMUS ,BRAIN tumors ,MTOR inhibitors ,MITOGEN-activated protein kinases ,GLIOMAS ,MAGNETIC resonance imaging ,CHILDREN'S hospitals - Abstract
Pediatric low-grade gliomas (pLGGs) are the most frequent brain tumor in children. Adjuvant treatment, consisting in chemotherapy and radiotherapy, is often necessary if a complete surgical resection cannot be obtained. Traditional treatment approaches result in a significant long-term morbidity, with a detrimental impact on quality of life. Dysregulation of the mitogen-activated protein kinase (MAPK) pathway is the molecular hallmark of pLGGs and hyperactivation of the downstream mammalian target of rapamycin (mTOR) pathway is frequently observed. We report clinical and radiological results of front-line treatment with everolimus in 10 consecutive patients diagnosed with m-TOR positive pLGGs at the Bambino Gesù Children's Hospital in Rome, Italy. Median duration of treatment was 19 months (range from 13-60). Brain magnetic resonance imaging showed stable disease in 7 patients, partial response in 1 and disease progression in 2. Therapy-related adverse events were always reversible after dose reduction or temporary treatment interruption. To the best of our knowledge, this is the first report of everolimus treatment for chemo- and radiotherapy-naïve children with pLGG. Our results provide preliminary support, despite low sample size, for the use of everolimus as target therapy in pLGG showing lack of progression with a manageable toxicity profile [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. A new baby in the c-Myc-directed transcriptional machinery: Che-1/AATF.
- Author
-
Folgiero, Valentina, Sorino, Cristina, Locatelli, Franco, and Fanciulli, Maurizio
- Published
- 2018
- Full Text
- View/download PDF
11. TIM-3/Gal-9 interaction induces IFNγ-dependent IDO1 expression in acute myeloid leukemia blast cells.
- Author
-
Folgiero, Valentina, Cifaldi, Loredana, Li Pira, Giuseppina, Goffredo, Bianca Maria, Vinti, Luciana, and Locatelli, Franco
- Subjects
- *
ACUTE myeloid leukemia , *GENE expression , *BONE marrow , *GENETIC regulation , *CANCER chemotherapy - Abstract
NK cells expressing TIM-3 show a marked increase in IFNγ production in response to acute myeloid leukemia (AML) blast cells that endogenously express Gal-9. Herein, we demonstrate that NK cell-mediated production of IFNγ, induced by TIM-3/Gal-9 interaction and released in bone marrow microenvironment, is responsible for IDO1 expression in AML blasts. IDO1-expressing AML blasts consequently down-regulate NK cell degranulation activity, by sustaining leukemia immune escape. Furthermore, the blocking of TIM-3/Gal-9 interaction strongly down-regulates IFNγ-dependent IDO1 activity. Thus, the inhibition of TIM-3/Gal-9 immune check point, which affects NK cell-dependent IFNγ production and the consequent IDO1 activation, could usefully integrate current chemotherapeutic approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
12. HIPK2 downregulates vimentin and inhibits breast cancer cell invasion.
- Author
-
Nodale, Cristina, Sheffer, Michal, Jacob-Hirsch, Jasmine, Folgiero, Valentina, Falcioni, Rita, Aiello, Aurora, Garufi, Alessia, Rechavi, Gideon, Givol, David, and D'Orazi, Gabriella
- Published
- 2012
- Full Text
- View/download PDF
13. Indoleamine 2,3-Dioxygenase-1 (IDO1) Is Expressed by a Subset of Childhood Acute Myeloid Leukemias and Restrains IFN-γ Production by T Cells
- Author
-
Folgiero, Valentina, Natale, Daniela, Pagliara, Daria, Caruso, Roberta, Vinti, Luciana, Coletti, Valentina, Moretta, Francesca, De Cristofaro, Raimondo, Locatelli, Franco, and Rutella, Sergio
- Published
- 2012
- Full Text
- View/download PDF
14. Induction of ErbB-3 Expression by α6β4 Integrin Contributes to Tamoxifen Resistance in ERβ1-Negative Breast Carcinomas.
- Author
-
Folgiero, Valentina, Avetrani, Paolo, Bon, Giulia, Di Carlo, Selene E., Fabi, Alessandra, Nisticò, Cecilia, Vici, Patrizia, Melucci, Elisa, Buglioni, Simonetta, Perracchio, Letizia, Sperduti, Isabella, Rosanò1, Laura, Sacchi, Ada, Mottolese, Marcella, and Falcioni, Rita
- Subjects
- *
PROTEIN kinases , *PROTEIN-tyrosine kinases , *TAMOXIFEN , *BREAST cancer treatment , *ADJUVANT treatment of cancer , *RANDOMIZED controlled trials , *ESTROGEN antagonists , *CANCER cells , *PHOSPHORYLATION - Abstract
Background: Tamoxifen is still the most widely used drug in hormone therapy for the treatment of breast cancer. Its benefits in adjuvant treatment are well documented in controlled and randomized clinical studies, which have demonstrated an increase in disease-free intervals of patients with positive hormonal receptors. However, the mechanisms involved in endocrine resistance are not clear. Laboratory and clinical data now indicate that bi-directional molecular cross-talk between nuclear or membrane ER and growth factor receptor pathways may be involved in endocrine resistance. We recently found a functional interaction between α6β4 integrin and ErbB-3 receptor to maintain the PI3K/Akt survival pathway of mammary tumour cells. We sought to improve understanding of this process in order to provide the involvement of both receptors insight into mechanism of Tamoxifen resistance. Methods and Findings: Using human breast cancer cell lines displaying different levels of α6β4 and ErbB-3 receptors and a series of 232 breast cancer biopsies from patients submitted to adjuvant Tamoxifen monotherapy for five years, we evaluated the functional interaction between both receptors in relationship to Tamoxifen responsiveness. In mammary carcinoma cells, we evidenced that the α6β4 integrin strongly influence Akt phosphorylation through ErbB-3 protein regulation. Moreover, the ErbB-3 inactivation inhibits Akt phosphorylation, induces apoptosis and inhibits in vitro invasion favouring Tamoxifen responsiveness. The analysis of human tumors revealed a significant relationship between α6β4 and ErbB-3 in P-Akt-positive and ERβ1-negative breast cancers derived from patients with lower disease free survival. Conclusions: We provided evidence that a strong relationship occurs between α6β4 and ErbB-3 positivity in ERβ1-negative breast cancers. We also found that the association between ErbB-3 and P-Akt positivity mainly occurs in ERβ1-negative breast cancer derived from patients with lower DFS indicating that both receptors are clinically relevant in predicting the response to Tamoxifen. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
15. Enhancement of Neuroblastoma NK-Cell-Mediated Lysis through NF-kB p65 Subunit-Induced Expression of FAS and PVR, the Loss of Which Is Associated with Poor Patient Outcome.
- Author
-
Brandetti, Elisa, Focaccetti, Chiara, Pezzolo, Annalisa, Ognibene, Marzia, Folgiero, Valentina, Cotugno, Nicola, Benvenuto, Monica, Palma, Paolo, Manzari, Vittorio, Rossi, Paolo, Fruci, Doriana, Bei, Roberto, and Cifaldi, Loredana
- Subjects
THERAPEUTIC use of interferons ,EVALUATION of medical care ,TISSUE arrays ,NEUROBLASTOMA ,KILLER cells ,CELL receptors ,APOPTOSIS ,GENE expression ,CANCER patients ,CELL survival ,DNA-binding proteins ,TUMOR necrosis factors ,CELL lines ,TUMOR markers ,CELL death ,IMMUNOTHERAPY - Abstract
Simple Summary: Neuroblastoma (NB) cells adopt several molecular strategies to evade the Natural Killer (NK)-mediated response. Herein, we found that the overexpression of the NF-kB p65 subunit in NB cell lines upregulates the expression of both the death receptor FAS and the activating ligand PVR, thus rendering NB cells more susceptible to NK-cell-mediated apoptosis, recognition, and killing. These data could provide a clue for a novel NK-cell-based immunotherapy of NB. In addition, array CGH analysis performed in our cohort of NB patients showed that loss of both the FAS and PVR genes correlated with low survival, thus revealing a novel biomarker predicting the outcome of NB patients. High-risk neuroblastoma (NB) is a rare childhood cancer whose aggressiveness is due to a variety of chromosomal genetic aberrations, including those conferring immune evasion. Indeed, NB cells adopt several molecular strategies to evade recognition by the immune system, including the downregulation of ligands for NK-cell-activating receptors. To date, while molecular strategies aimed at enhancing the expression of ligands for NKG2D- and DNAM-1-activating receptors have been explored, no evidence has been reported on the immunomodulatory mechanisms acting on the expression of death receptors such as Fas in NB cells. Here, we demonstrated that transient overexpression of the NF-kB p65 subunit upregulates the surface expression of Fas and PVR, the ligand of DNAM-1, thus making NB cell lines significantly more susceptible to NK-cell-mediated apoptosis, recognition, and killing. In contrast, IFNγ and TNFα treatment, although it induced the upregulation of FAS in NB cells and consequently enhanced NK-cell-mediated apoptosis, triggered immune evasion processes, including the strong upregulation of MHC class I and IDO1, both of which are involved in mechanisms leading to the impairment of a proper NK-cell-mediated killing of NB. In addition, high-resolution array CGH analysis performed in our cohort of NB patients revealed that the loss of FAS and/or PVR genes correlated with low survival independently of the disease stage. Our data identify the status of the FAS and PVR genes as prognostic biomarkers of NB that may predict the efficacy of NK-cell-based immunotherapy of NB. Overall, restoration of surface expression of Fas and PVR, through transient upregulation of NF-kB, may be a clue to a novel NK-cell-based immunotherapy of NB. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. PI3K/Akt Pathway: The Indestructible Role of a Vintage Target as a Support to the Most Recent Immunotherapeutic Approaches.
- Author
-
Caforio, Matteo, de Billy, Emmanuel, De Angelis, Biagio, Iacovelli, Stefano, Quintarelli, Concetta, Paganelli, Valeria, and Folgiero, Valentina
- Subjects
PHOSPHOTRANSFERASES ,CELL physiology ,IMMUNE system ,ANTINEOPLASTIC agents ,CELLULAR signal transduction ,IMMUNOLOGICAL adjuvants ,TRANSFERASES ,TUMOR markers ,TUMORS ,IMMUNOTHERAPY - Abstract
Simple Summary: PI3K/Akt pathway has an impressive story as tumor marker. PI3K-dependent solid tumors have been studied for several years in order to inhibit the pathway at different levels along the signaling. Despite the highly satisfactory results obtained in vitro and in xenograft mouse tumor models, the use of PI3K/Akt inhibitors in clinical trials resulted in being not as efficient as expected. With the emerging role of the tumor microenvironment in the response to therapy and the awareness, increasing in recent years, of the necessity to army the immune system against the tumor, new opportunities have emerged for PI3K/Akt inhibitors. Here, we show that PI3K/Akt, in addition to its function as tumor marker, exerts a pivotal role as an immunomodulator. Recent studies demonstrate that PI3K/Akt pathway is crucial for the regulation of the immune system and that its inhibition in combination with immunomodulatory agents may provide a new therapeutic approach for cancer. Pathologic activation of PI3Ks and the subsequent deregulation of its downstream signaling pathway is among the most frequent events associated with cellular transformation, cancer, and metastasis. PI3Ks are also emerging as critical factors in regulating anti-tumor immunity by either promoting an immunosuppressive tumor microenvironment or by controlling the activity and the tumor infiltration of cells involved in the immune response. For these reasons, significant pharmaceutical efforts are dedicated to inhibiting the PI3K pathway, with the main goal to target the tumor and, at the same time, to enhance the anti-tumor immunity. Recent immunotherapeutic approaches involving the use of adoptive cell transfer of autologous genetically modified T cells or immune check-point inhibitors showed high efficacy. However, mechanisms of resistance to these kinds of therapy are emerging, due in part to the inhibition of effector T cell functions exerted by the immunosuppressive tumor microenvironment. Here, we first describe how inhibition of PI3K/Akt pathway contribute to enhance anti-tumor immunity and further discuss how inhibitors of the pathway are used in combination with different immunomodulatory and immunotherapeutic agents to improve anti-tumor efficacy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. Proteomics Study of Peripheral Blood Mononuclear Cells in Down Syndrome Children.
- Author
-
Lanzillotta, Chiara, Greco, Viviana, Valentini, Diletta, Villani, Alberto, Folgiero, Valentina, Caforio, Matteo, Locatelli, Franco, Pagnotta, Sara, Barone, Eugenio, Urbani, Andrea, Di Domenico, Fabio, and Perluigi, Marzia
- Subjects
DOWN syndrome ,BLOOD cells ,PROTEOMICS ,CYTOSKELETON ,WESTERN immunoblotting ,DNA structure - Abstract
Down syndrome (DS) is the most common chromosomal disorder and the leading genetic cause of intellectual disability in humans, which results from the triplication of chromosome 21. To search for biomarkers for the early detection and exploration of the disease mechanisms, here, we investigated the protein expression signature of peripheral blood mononuclear cells (PBMCs) in DS children compared with healthy donors (HD) by using an in-depth label-free shotgun proteomics approach. Identified proteins are found associated with metabolic pathways, cellular trafficking, DNA structure, stress response, cytoskeleton network, and signaling pathways. The results showed that a well-defined number of dysregulated pathways retain a prominent role in mediating DS pathological features. Further, proteomics results are consistent with published study in DS and provide evidences that increased oxidative stress and the increased induction of stress related response, is a participant in DS pathology. In addition, the expression levels of some key proteins have been validated by Western blot analysis while protein carbonylation, as marker of protein oxidation, was investigated. The results of this study propose that PBMCs from DS children might be in an activated state where endoplasmic reticulum stress and increased production of radical species are one of the primary events contributing to multiple DS pathological features. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
18. CAPE and its synthetic derivative VP961 restore BACH1/NRF2 axis in Down Syndrome.
- Author
-
Pagnotta, Sara, Tramutola, Antonella, Barone, Eugenio, Di Domenico, Fabio, Pittalà, Valeria, Salerno, Loredana, Folgiero, Valentina, Caforio, Matteo, Locatelli, Franco, Petrini, Stefania, Butterfield, D. Allan, and Perluigi, Marzia
- Subjects
- *
DOWN syndrome , *LYMPHOBLASTOID cell lines , *REACTIVE nitrogen species , *HEME oxygenase , *SUPEROXIDE dismutase , *TRANSCRIPTION factors - Abstract
The cells possess several mechanisms to counteract the over-production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), including enzymes such as superoxide dismutase, catalase and glutathione peroxidase. Moreover, an important sensor involved in the anti-oxidant response is KEAP1-NRF2-ARE signaling complex. Under oxidative stress (OS), the transcription factor NRF2 can dissociate from the KEAP1-complex in the cytosol and translocate into the nucleus to promote the transcriptional activation of anti-oxidant genes, such as heme oxygenase 1 and NADPH quinone oxidoreductase. Within this context, the activation of NRF2 response is further regulated by BACH1, a transcription repressor, that compete with the KEAP1-NRF2-ARE complex. In this work, we focused on the role of BACH1/NRF2 ratio in the regulation of the anti-oxidant response, proposing their antithetical relation as a valuable target for a therapeutic strategy to test drugs able to exert neuroprotective effects, notably in aging and neurodegenerative diseases. Among these, Down syndrome (DS) is a complex genetic disorder characterized by BACH1 gene triplication that likely results in the impairment of NRF2 causing increased OS. Our results revealed that BACH1 overexpression alters the BACH1/NRF2 ratio in the nucleus and disturbs the induction of antioxidant response genes ultimately resulting in the accumulation of oxidative damage both in Ts2Cje mice (a mouse model of DS) and human DS lymphoblastoid cell lines (LCLs). Based on this evidence, we tested Caffeic Acid Phenethyl Ester (CAPE) and the synthetic analogue VP961, which have been proven to modulate NRF2 activity. We showed that CAPE and VP961 administration to DS LCLs was able to promote NRF2 nuclear translocation, which resulted in the amelioration of antioxidant response. Overall, our study supports the hypothesis that BACH1 triplication in DS subjects is implicated in the alteration of redox homeostasis and therapeutic strategies to overcome this effect are under investigation in our laboratory. [Display omitted] • BACH-1/Nrf-2 has a fundamental role in the regulation of anti-oxidant response. • BACH-1 is among the genes encoded on HSA21 found to be overexpressed in Down Syndrome. • Targeting BACH-1/Nrf-2 axis is a valid therapeutic strategy in disease with increased Oxidative Stress. • Oxidative stress is an early event in Down Syndrome. • CAPE and VP961 are able to restore BACH-1/Nrf-2 axis in Down Syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
19. Chronic PERK induction promotes Alzheimer-like neuropathology in Down syndrome: Insights for therapeutic intervention.
- Author
-
Lanzillotta, Chiara, Zuliani, Ilaria, Tramutola, Antonella, Barone, Eugenio, Blarzino, Carla, Folgiero, Valentina, Caforio, Matteo, Valentini, Diletta, Villani, Alberto, Locatelli, Franco, Butterfield, D. Allan, Head, Elizabeth, Perluigi, Marzia, Abisambra, Jose F., and Di Domenico, Fabio
- Subjects
- *
UNFOLDED protein response , *DOWN syndrome , *NEUROLOGICAL disorders , *NEURAL development , *PROTEIN synthesis - Abstract
• The PERK branch of the unfolded protein response is induced in DS and AD patients • Over-induced PERK increases eIF2α deregulation promoting translation repression • DS brain demonstrates the uncoupling between PERK and Nrf2-related antioxidant response • The inhibition of PERK in DS mice rescue proteins translation through the reduction of eIF2α • The inhibition of PERK rebalances Nrf2/Bach1 nuclear levels and decrease brain oxidative damage A major challenge in neurobiology is the identification of the mechanisms by which protein misfolding leads to cellular toxicity. Many neurodegenerative disorders, in which aberrant protein conformers aggregate into pathological inclusions, present the chronic activation of the PERK branch of the unfolded protein response. The adaptive effects of the PERK pathway include reduction of translation by transient inhibition of eIF2α and antioxidant protein production via induction of Nrf2 transcription factor. In contrast, PERK prolonged activation leads to sustained reduction in protein synthesis and induction of cell death pathways. To further investigate the role of the PERK pathway in neurodegenerative disorders, we focused on Down syndrome (DS), in which aging confers a high risk of Alzheimer disease (AD). By investigating human DS frontal cortices, we found early and sustained PERK activation associated with the induction of eIF2α and ATF4 downstream signals. We also observed that the Nrf2 response is uncoupled from PERK and its antioxidant effects are repressed in a mechanism implicating the transcription repressor Bach1. The pharmacological inhibition of PERK in DS mice reduced eIF2α-related translational repression and promoted Nrf2 nuclear translocation, favoring the rescue of Nrf2/Bach1 imbalance. The further analysis of peripheral cells from living DS individuals provided strong support of the pathological link between PERK and trisomy 21. Our results suggest that failure to regulate the PERK pathway is a peculiar characteristic of DS pathology and it may represent an essential step to promote cellular dysfunction, which actively contributes in the brain to the early development of AD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
20. GD2 redirected CAR T and activated NK-cell-mediated secretion of IFNγ overcomes MYCN-dependent IDO1 inhibition, contributing to neuroblastoma cell immune escape.
- Author
-
Caforio M, Sorino C, Caruana I, Weber G, Camera A, Cifaldi L, De Angelis B, Del Bufalo F, Vitale A, Goffredo BM, De Vito R, Fruci D, Quintarelli C, Fanciulli M, Locatelli F, and Folgiero V
- Subjects
- Cell Line, Tumor, Coculture Techniques, Gangliosides immunology, Gene Expression Regulation, Neoplastic, Humans, Indoleamine-Pyrrole 2,3,-Dioxygenase genetics, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, N-Myc Proto-Oncogene Protein genetics, Neuroblastoma enzymology, Neuroblastoma genetics, Neuroblastoma immunology, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Signal Transduction, T-Lymphocytes immunology, T-Lymphocytes metabolism, Tumor Escape, Tumor Microenvironment, Gangliosides metabolism, Immunotherapy, Adoptive, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Interferon-gamma metabolism, Killer Cells, Natural transplantation, Lymphocyte Activation, N-Myc Proto-Oncogene Protein metabolism, Neuroblastoma therapy, Receptors, Chimeric Antigen genetics, T-Lymphocytes transplantation
- Abstract
Immune escape mechanisms employed by neuroblastoma (NB) cells include secretion of immunosuppressive factors disrupting effective antitumor immunity. The use of cellular therapy to treat solid tumors needs to be implemented. Killing activity of anti-GD2 Chimeric Antigen Receptor (CAR) T or natural killer (NK) cells against target NB cells was assessed through coculture experiments and quantified by FACS analysis. ELISA assay was used to quantify interferon-γ (IFNγ) secreted by NK and CAR T cells. Real Time PCR and Western Blot were performed to analyze gene and protein levels modifications. Transcriptional study was performed by chromatin immunoprecipitation and luciferase reporter assays on experiments of mutagenesis on the promoter sequence. NB tissue sample were analyzed by IHC and Real Time PCR to perform correlation study. We demonstrate that Indoleamine-pyrrole 2,3-dioxygenase1 (IDO1), due to its ability to convert tryptophan into kynurenines, is involved in NB resistance to activity of immune cells. In NB, IDO1 is able to inhibit the anti-tumor effect displayed by of both anti-GD2 CAR (GD2.CAR) T-cell and NK cells, mainly by impairing their IFNγ production. Furthermore, inhibition of MYCN expression in NB results into accumulation of IDO1 and consequently of kynurenines, which negatively affect the immune surveillance. Inverse correlation between IDO1 and MYCN expression has been observed in a wide cohort of NB samples. This finding was supported by the identification of a transcriptional repressive role of MYCN on IDO1 promoter. The evidence of IDO1 involvement in NB immune escape and its ability to impair NK and GD2.CAR T-cell activity contribute to clarify one of the possible mechanisms responsible for the limited efficacy of these immunotherapeutic approaches. A combined therapy of NK or GD2.CAR T-cells with IDO1 inhibitors, a class of compounds already in phase I/II clinical studies, could represent a new and still unexplored strategy capable to improve long-term efficacy of these immunotherapeutic approaches., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
21. IDO1 involvement in mTOR pathway: a molecular mechanism of resistance to mTOR targeting in medulloblastoma.
- Author
-
Folgiero V, Miele E, Carai A, Ferretti E, Alfano V, Po A, Bertaina V, Goffredo BM, Benedetti MC, Camassei FD, Cacchione A, Locatelli F, and Mastronuzzi A
- Subjects
- Antibiotics, Antineoplastic therapeutic use, Cell Line, Tumor, Cerebellar Neoplasms drug therapy, Cerebellar Neoplasms metabolism, Child, Gene Expression Regulation, Neoplastic drug effects, Humans, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Infant, Medulloblastoma drug therapy, Medulloblastoma metabolism, Molecular Targeted Therapy, Signal Transduction drug effects, Sirolimus therapeutic use, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, TOR Serine-Threonine Kinases metabolism, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Cerebellar Neoplasms genetics, Drug Resistance, Neoplasm genetics, Indoleamine-Pyrrole 2,3,-Dioxygenase genetics, Medulloblastoma genetics, Signal Transduction genetics, TOR Serine-Threonine Kinases genetics
- Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. Despite therapeutic advancements, high-risk groups still present significant mortality. A deeper knowledge of the signaling pathways contributing to MB formation and aggressiveness would help develop new successful therapies. The target of rapamycin, mTOR signaling, is known to be involved in MB and is already targetable in the clinical setting. Furthermore, mTOR is a master metabolic regulator able to control cell growth versus autophagy decisions in conditions of amino-acid deprivation that can be due to IDO1 enzymatic activity. IDO1 has been also implicated in the regulation of inflammation, as well as of T cell-mediated immune responses, in a variety of pathological conditions, including brain tumors. In particular, IDO1 induces expansion of regulatory T-cells (Treg), preventing immune response against tumor cells. Analysis of 27 MB tissue specimens for the expression of both mTOR and IDO1 showed their widespread expression in all samples. Testing their cooperation in vitro, a significant involvement of IDO1 in mTOR immunogenic pathway was found, able to counteract the aim of rapamycin treatment. In MB cell lines, inhibition of mTOR strongly induced IDO1 expression and activity, corroborating its ability to recruit Treg cells in the tumor microenvironment. The mTOR/IDO1 cross talk was found to be strictly specific of MB cells. We demonstrated that mTOR pathway cross talks with IDO1 pathway to promote MB immune escape, possibly contributing to failure of mTOR- targeted therapy., Competing Interests: None of the Authors has any conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
22. Indoleamine 2,3-dioxygenase 1 (IDO1) activity in leukemia blasts correlates with poor outcome in childhood acute myeloid leukemia.
- Author
-
Folgiero V, Goffredo BM, Filippini P, Masetti R, Bonanno G, Caruso R, Bertaina V, Mastronuzzi A, Gaspari S, Zecca M, Torelli GF, Testi AM, Pession A, Locatelli F, and Rutella S
- Subjects
- Adolescent, Blotting, Western, Child, Child, Preschool, Disease-Free Survival, Enzyme-Linked Immunosorbent Assay, Female, Fluorescent Antibody Technique, Humans, Immunoprecipitation, Infant, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Male, Real-Time Polymerase Chain Reaction, Young Adult, Biomarkers, Tumor analysis, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Leukemia, Myeloid, Acute enzymology
- Abstract
Microenvironmental factors contribute to the immune dysfunction characterizing acute myeloid leukemia (AML). Indoleamine 2,3-dioxygenase 1 (IDO1) is an interferon (IFN)-γ-inducible enzyme that degrades tryptophan into kynurenine, which, in turn, inhibits effector T cells and promotes regulatory T-cell (Treg) differentiation. It is presently unknown whether childhood AML cells express IDO1 and whether IDO1 activity correlates with patient outcome. We investigated IDO1 expression and function in 37 children with newly diagnosed AML other than acute promyelocytic leukemia. Blast cells were cultured with exogenous IFN-γ for 24 hours, followed by the measurement of kynurenine production and tryptophan consumption. No constitutive expression of IDO1 protein was detected in blast cells from the 37 AML samples herein tested. Conversely, 19 out of 37 (51%) AML samples up-regulated functional IDO1 protein in response to IFN-γ. The inability to express IDO1 by the remaining 18 AML samples was not apparently due to a defective IFN-γ signaling circuitry, as suggested by the measurement of signal transducer and activator of transcription 3 (STAT3) phosphorylation. Co-immunoprecipitation assays indicated the occurrence of physical interactions between STAT3 and IDO1 in AML blasts. In line with this finding, STAT3 inhibitors abrogated IDO1 function in AML blasts. Interestingly, levels of IFN-γ were significantly higher in the bone marrow fluid of IDO-expressing compared with IDO-nonexpressing AMLs. In mixed tumor lymphocyte cultures (MTLC), IDO-expressing AML blasts blunted the ability of allogeneic naïve T cells to produce IFN-γ and promoted Treg differentiation. From a clinical perspective, the 8-year event-free survival was significantly worse in IDO-expressing children (16.4%, SE 9.8) as compared with IDO-nonexpressing ones (48.0%, SE 12.1; p=0.035). These data indicate that IDO1 expression by leukemia blasts negatively affects the prognosis of childhood AML. Moreover, they speak in favor of the hypothesis that IDO can be targeted, in adjunct to current chemotherapy approaches, to improve the clinical outcome of children with AML.
- Published
- 2014
- Full Text
- View/download PDF
23. Transcription factor NF-Y induces apoptosis in cells expressing wild-type p53 through E2F1 upregulation and p53 activation.
- Author
-
Gurtner A, Fuschi P, Martelli F, Manni I, Artuso S, Simonte G, Ambrosino V, Antonini A, Folgiero V, Falcioni R, Sacchi A, and Piaggio G
- Subjects
- Animals, Blotting, Western, CCAAT-Binding Factor genetics, CCAAT-Binding Factor metabolism, Cell Line, Cell Line, Tumor, Cell Proliferation, Cells, Cultured, E2F1 Transcription Factor genetics, Embryo, Mammalian cytology, Fibroblasts cytology, Fibroblasts metabolism, HCT116 Cells, HeLa Cells, Humans, Mice, Mice, Knockout, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction physiology, Tumor Suppressor Protein p53 genetics, Apoptosis physiology, CCAAT-Binding Factor physiology, E2F1 Transcription Factor metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The CCAAT-binding transcription factor NF-Y plays a central role in regulating cellular proliferation by controlling the expression of genes required for cell-cycle progression such as cyclin A, cyclin B1, cyclin B2, cdc25A, cdc25C, and cdk1. Here we show that unrestricted NF-Y activity leads to apoptosis in an E2F1- and wild-type p53 (wtp53)-dependent manner. Unrestricted NF-Y activity induced an increase in E2F1 mRNA and protein levels. Furthermore, NF-Y directly bound the E2F1 promoter and this correlated with the appearance of open chromatin marks. The ability of NF-Y to induce apoptosis was impaired in cells lacking E2F1 and wtp53. Moreover, NF-Y overexpression elicited phosphorylation of wt p53Ser18 in an E2F1-dependent manner. Our findings establish that NF-Y acts upstream of E2F1 in p53-mediated apoptosis.
- Published
- 2010
- Full Text
- View/download PDF
24. Purification and characterization of adipose-derived stem cells from patients with lipoaspirate transplant.
- Author
-
Folgiero V, Migliano E, Tedesco M, Iacovelli S, Bon G, Torre ML, Sacchi A, Marazzi M, Bucher S, and Falcioni R
- Subjects
- Adipocytes metabolism, Adipose Tissue cytology, Adult, Adult Stem Cells metabolism, Aged, Biomarkers metabolism, Cell Differentiation, Cells, Cultured, Flow Cytometry, Humans, Membrane Proteins metabolism, Membrane Proteins physiology, Middle Aged, Tissue and Organ Harvesting, Adipocytes cytology, Adult Stem Cells cytology
- Abstract
Techniques for medical tissue regeneration require an abundant source of human adult stem cells. There is increasing evidence that adipose stem cells contribute to restoration of tissue vascularization and organ function. The object of our study was to isolate and characterize adult adipose-derived stem cells from patients undergoing on lipoaspirate transplant with the aim to improve tissue regeneration. Adipose-derived stem cells were isolated and purified from the lipoaspirate of 15 patients and characterized for CD markers and the ability to differentiate toward the adipogenic lineage. We found that purified adipose stem cells express high level of CD49d, CD44, CD90, CD105, CD13, and CD71 and these markers of staminality were maintained at high level for at least 3 months and seven passages of in vitro culture. As expected, these cells resulted negative for the endothelial and hematopoietic-specific markers CD31, CD106, CD34, and CD45. Differentiation towards adipogenic lineage demonstrated that purified adipose-derived stem cells are still able to become adipocytes at least 3 months after in vitro culture. The analysis of Akt and MAPK phosphorylation confirmed a modulation of their activity during differentiation. Interestingly, we established for the first time that, among the p53 family members, a strong upregulation of p63 expression occurs in adipocytic differentiation, indicating a role for this transcription factor in adipocytic differentiation. Taken together, these data indicate that purified lipoaspirate-derived stem cells maintain their characteristic of staminality for a long period of in vitro culture, suggesting that they could be applied for cell-based therapy to improve autologous lipoaspirate transplant.
- Published
- 2010
- Full Text
- View/download PDF
25. Negative regulation of beta4 integrin transcription by homeodomain-interacting protein kinase 2 and p53 impairs tumor progression.
- Author
-
Bon G, Di Carlo SE, Folgiero V, Avetrani P, Lazzari C, D'Orazi G, Brizzi MF, Sacchi A, Soddu S, Blandino G, Mottolese M, and Falcioni R
- Subjects
- Blotting, Western, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carrier Proteins genetics, Cell Line, Tumor, Cell Proliferation, Cytoplasm metabolism, Disease Progression, Female, Gene Expression Regulation, Neoplastic, HCT116 Cells, HT29 Cells, Histone Acetyltransferases metabolism, Humans, Immunohistochemistry, Integrin beta4 genetics, Mitogen-Activated Protein Kinases metabolism, Mutation, Neoplasms genetics, Neoplasms metabolism, Phosphorylation, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins c-akt metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic, Tumor Suppressor Protein p53 genetics, p300-CBP Transcription Factors metabolism, Carrier Proteins metabolism, Integrin beta4 metabolism, Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Increased expression of alpha(6)beta(4) integrin in several epithelial cancers promotes tumor progression; however, the mechanism underlying its transcriptional regulation remains unclear. Here, we show that depletion of homeodomain-interacting protein kinase 2 (HIPK2) activates beta(4) transcription that results in a strong increase of beta(4)-dependent mitogen-activated protein kinase and Akt phosphorylation, anchorage-independent growth, and invasion. In contrast, stabilization of HIPK2 represses beta(4) expression in wild-type p53 (wtp53)-expressing cells but not in p53-null cells or cells expressing mutant p53, indicating that HIPK2 requires a wtp53 to inhibit beta(4) transcription. Consistent with our in vitro findings, a strong correlation between beta(4) overexpression and HIPK2 inactivation by cytoplasmic relocalization was observed in wtp53-expressing human breast carcinomas. Under loss of function of HIPK2 or p53, the p53 family members TAp63 and TAp73 strongly activate beta(4) transcription. These data, by revealing that beta(4) expression is transcriptionally repressed in tumors by HIPK2 and p53 to impair beta(4)-dependent tumor progression, suggest that loss of p53 function favors the formation of coactivator complex with the TA members of the p53 family to allow beta(4) transcription.
- Published
- 2009
- Full Text
- View/download PDF
26. The alpha6beta4 integrin can regulate ErbB-3 expression: implications for alpha6beta4 signaling and function.
- Author
-
Folgiero V, Bachelder RE, Bon G, Sacchi A, Falcioni R, and Mercurio AM
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Carcinoma genetics, Carcinoma metabolism, Cell Line, Tumor, Dimerization, Enzyme Activation, Eukaryotic Initiation Factor-4F metabolism, Humans, Integrin alpha6beta4 biosynthesis, Mice, NIH 3T3 Cells, Neuregulin-1 pharmacology, Phosphatidylinositol 3-Kinases metabolism, Protein Biosynthesis, Proto-Oncogene Proteins c-akt metabolism, RNA, Messenger biosynthesis, RNA, Messenger genetics, Receptor, ErbB-2 metabolism, Receptor, ErbB-3 genetics, Signal Transduction, Integrin alpha6beta4 metabolism, Receptor, ErbB-3 biosynthesis
- Abstract
The integrin alpha(6)beta(4) has been shown to facilitate key functions of carcinoma cells, including their ability to migrate, invade, and evade apoptosis. The mechanism involved seems to be a profound effect of alpha(6)beta(4) on specific signaling pathways, especially the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. An intimate relationship between alpha(6)beta(4) and growth factor receptors may explain this effect of alpha(6)beta(4) on signaling. Previously, we showed that alpha(6)beta(4) and ErbB-2 can function synergistically to activate the PI3K/Akt pathway. Given that ErbB-2 can activate PI3K only when it heterodimerizes with other members of the epidermal growth factor receptor family, these data imply that other receptors cooperate in this process. Here, we report that alpha(6)beta(4) can regulate the expression of ErbB-3 using several different models and that the consequent formation of an ErbB-2/ErbB-3 heterodimer promotes the alpha(6)beta(4)-dependent activation of PI3K/Akt and the ability of this integrin to impede apoptosis of carcinoma cells. Our data also support the hypothesis that alpha(6)beta(4) can regulate ErbB-3 expression at the translational level as evidenced by the findings that alpha(6)beta(4) does not increase ErbB-3 mRNA significantly, and that this regulation is both rapamycin sensitive and dependent on eukaryotic translation initiation factor 4E. These findings provide one mechanism to account for the activation of PI3K by alpha(6)beta(4) and they also provide insight into the regulation of ErbB-3 in carcinoma cells.
- Published
- 2007
- Full Text
- View/download PDF
27. Involvement of alpha6beta4 integrin in the mechanisms that regulate breast cancer progression.
- Author
-
Bon G, Folgiero V, Di Carlo S, Sacchi A, and Falcioni R
- Subjects
- Cell Movement, Cell Survival, Disease Progression, Female, Humans, Signal Transduction, Breast Neoplasms pathology, Carcinoma pathology, Integrin alpha6beta4 physiology
- Abstract
Integrin alpha6beta4 is mostly expressed in epithelial tissues and endothelial and Schwann cells. Expression of alpha6beta4 is increased in many epithelial tumours, implicating its involvement in tumour malignancy. Moreover, this integrin activates several key signalling molecules in carcinoma cells, but its ability to activate the phosphatidylinositol 3-kinase/Akt pathway is among the mechanisms by which alpha6beta4 integrin regulates tumour behaviour. In this review we discuss the biological and clinical features of alpha6beta4 integrin that allow it to promote tumour survival and progression of mammary tumours.
- Published
- 2007
- Full Text
- View/download PDF
28. Loss of beta4 integrin subunit reduces the tumorigenicity of MCF7 mammary cells and causes apoptosis upon hormone deprivation.
- Author
-
Bon G, Folgiero V, Bossi G, Felicioni L, Marchetti A, Sacchi A, and Falcioni R
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation drug effects, Female, Gene Expression Profiling, Humans, Integrin beta4 drug effects, Integrin beta4 genetics, Mice, Mice, Nude, Phosphatidylinositol 3-Kinases drug effects, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Protein Kinases drug effects, Protein Kinases metabolism, Protein Subunits drug effects, Protein Subunits genetics, Protein Subunits metabolism, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering metabolism, RNA, Small Interfering pharmacology, Reverse Transcriptase Polymerase Chain Reaction methods, Signal Transduction drug effects, Structure-Activity Relationship, TOR Serine-Threonine Kinases, Tamoxifen pharmacology, Xenograft Model Antitumor Assays, Apoptosis, Breast Neoplasms metabolism, Integrin beta4 metabolism
- Abstract
Purpose: The alpha6beta4 integrin, a laminin receptor, has been implicated from many studies in tumor progression and invasion. We showed that the beta4 integrin subunit associates with the ErbB-2 tyrosine kinase in human mammary carcinoma cell lines and that its overexpression in NIH3T3/ErbB-2-transformed cells causes a constitutive activation of phosphatidylinositol 3-kinase (PI3K), inducing a strong increase of their invasive capacity. In this study, we investigated the biological consequences of interference with the endogenous beta4 integrin subunit expression., Experimental Design: In vitro and in vivo tumor growth and the biochemical consequences of beta4 integrin inactivation were studied in mammary tumor cells by using short hairpin RNA approach., Results: Our data show that tumor growth of mammary tumor cells strictly depends on beta4 expression, confirming the relevance of beta4 protein in these cells. Moreover, interference with beta4 expression significantly reduces endogenous PI3K activity and AKT and mammalian target of rapamycin phosphorylation. Accordingly, with these results and considering that PI3K activity in mammary tumor plays a relevant role in hormone resistance, we asked whether beta4 expression might be relevant for hormone responsiveness in these cells. Data reported indicate that the interference with endogenous beta4 expression, upon hormone deprivation, induces caspase-9 and cytochrome c-mediated apoptosis, which is enhanced upon tamoxifen treatment. On the other hand, the expression of myr-AKT in MCF7 beta4-short hairpin RNA cells rescues the cells from apoptosis in the absence of hormones and upon tamoxifen treatment., Conclusions: Overall, these results confirm the relevance of beta4 expression in mammary tumors and indicate this integrin as a relevant target for tumor therapy.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.