Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marco Genua is active.

Publication


Featured researches published by Marco Genua.


Journal of Biological Chemistry | 2011

Decorin Antagonizes IGF Receptor I (IGF-IR) Function by Interfering with IGF-IR Activity and Attenuating Downstream Signaling

Renato V. Iozzo; Simone Buraschi; Marco Genua; Shi-Qiong Xu; Charalambos Solomides; Stephen C. Peiper; Leonard G. Gomella; Rick C. Owens; Andrea Morrione

We have recently discovered that the insulin-like growth factor receptor I (IGF-IR) is up-regulated in human invasive bladder cancer and promotes migration and invasion of transformed urothelial cells. The proteoglycan decorin, a key component of the tumor stroma, can positively regulate the IGF-IR system in normal cells. However, there are no available data on the role of decorin in modulating IGF-IR activity in transformed cells or in tumor models. Here we show that the expression of decorin inversely correlated with IGF-IR expression in low and high grade bladder cancers (n = 20 each). Decorin bound with high affinity IGF-IR and IGF-I at distinct sites and negatively regulated IGF-IR activity in urothelial cancer cells. Nanomolar concentrations of decorin promoted down-regulation of IRS-1, one of the critical proteins of the IGF-IR pathway, and attenuated IGF-I-dependent activation of Akt and MAPK. This led to decorin-evoked inhibition of migration and invasion upon IGF-I stimulation. Notably, decorin did not cause down-regulation of the IGF-IR in bladder, breast, and squamous carcinoma cells. This indicates that decorin action on the IGF-IR differs from its known activity on other receptor tyrosine kinases such as the EGF receptor and Met. Our results provide a novel mechanism for decorin in negatively modulating both IGF-I and its receptor. Thus, decorin loss may contribute to increased IGF-IR activity in the progression of bladder cancer and perhaps other forms of cancer where IGF-IR plays a role.


American Journal of Pathology | 2010

The insulin-like growth factor receptor I promotes motility and invasion of bladder cancer cells through Akt- and mitogen-activated protein kinase-dependent activation of paxillin.

David Metalli; Francesca Lovat; Farida Tripodi; Marco Genua; Shi-Qiong Xu; Michela Spinelli; Lilia Alberghina; Marco Vanoni; Raffaele Baffa; Leonard G. Gomella; Renato V. Iozzo; Andrea Morrione

The insulin-like growth factor receptor I (IGF-IR) plays an essential role in transformation by promoting cell growth and protecting cancer cells from apoptosis. Aberrant IGF-IR signaling is implicated in several types of tumors, including carcinomas of the lung, breast, prostate, pancreas, liver, and colon. However, the contribution of the IGF-IR to the development of the transformed phenotype in urothelial cells has not been clearly established. In this study we demonstrated that the IGF-IR is overexpressed in invasive bladder cancer tissues compared with nonmalignant controls. We have investigated the role of the IGF-IR in bladder cancer by using urothelial carcinoma-derived 5637 and T24 cells. Although activation of the IGF-IR did not appreciably affect their growth, it did promote migration and stimulate in vitro wound closure and invasion. These effects required the activation of the Akt and Mitogen-activated protein kinase (MAPK) pathways as well as IGF-I-induced Akt- and MAPK-dependent phosphorylation of paxillin, which relocated at dynamic focal adhesions and was necessary for promoting motility in bladder cancer cells. Our results provide the first evidence for a role of the IGF-IR in motility and invasion of bladder cancer cells and support the hypothesis that the IGF-IR may play a critical role in the establishment of the invasive phenotype in urothelial neoplasia. Thus, the IGF-IR may also serve as a novel biomarker for bladder cancer.


Journal of Biological Chemistry | 2012

Insulin and Insulin-like Growth Factor II Differentially Regulate Endocytic Sorting and Stability of Insulin Receptor Isoform A

Alaide Morcavallo; Marco Genua; Angela Palummo; Emília Kletvíková; Jiri Jiracek; Andrzej M. Brzozowski; Renato V. Iozzo; Antonino Belfiore; Andrea Morrione

Background: Insulin-like growth factor (IGF) II interacts with IR-A and is a more powerful mitogen than insulin. Results: IGF-II and insulin differ in regulating insulin receptor (IR)-A trafficking and stability. Conclusion: Compared with insulin, IGF-II induces lower IR-A and downstream effectors activation but protects IR-A and IRS-1 from down-regulation, thereby evoking a sustained mitogenic stimulus. Significance: These results further elucidate the mechanisms controlling IR-A biological responses. The insulin receptor isoform A (IR-A) binds both insulin and insulin-like growth factor (IGF)-II, although the affinity for IGF-II is 3–10-fold lower than insulin depending on a cell and tissue context. Notably, in mouse embryonic fibroblasts lacking the IGF-IR and expressing solely the IR-A (R−/IR-A), IGF-II is a more potent mitogen than insulin. As receptor endocytosis and degradation provide spatial and temporal regulation of signaling events, we hypothesized that insulin and IGF-II could affect IR-A biological responses by differentially regulating IR-A trafficking. Using R−/IR-A cells, we discovered that insulin evoked significant IR-A internalization, a process modestly affected by IGF-II. However, the differential internalization was not due to IR-A ubiquitination. Notably, prolonged stimulation of R−/IR-A cells with insulin, but not with IGF-II, targeted the receptor to a degradative pathway. Similarly, the docking protein insulin receptor substrate 1 (IRS-1) was down-regulated after prolonged insulin but not IGF-II exposure. Similar results were also obtained in experiments using [NMeTyrB26]-insulin, an insulin analog with IR-A binding affinity similar to IGF-II. Finally, we discovered that IR-A was internalized through clathrin-dependent and -independent pathways, which differentially regulated the activation of downstream effectors. Collectively, our results suggest that a lower affinity of IGF-II for the IR-A promotes lower IR-A phosphorylation and activation of early downstream effectors vis à vis insulin but may protect IR-A and IRS-1 from down-regulation thereby evoking sustained and robust mitogenic stimuli.


Cancer Research | 2007

17β-Estradiol Up-regulates the Insulin-like Growth Factor Receptor through a Nongenotropic Pathway in Prostate Cancer Cells

Giuseppe Pandini; Marco Genua; Francesco Frasca; Sebastiano Squatrito; Riccardo Vigneri; Antonino Belfiore

Prostate carcinomas frequently express estrogen receptors (ER), irrespective of androgen receptor (AR) expression; however, the role of ERs and estrogens in prostate cancer is controversial. We found that 17β-estradiol (E 2 ) is able to markedly up-regulate insulin-like growth factor (IGF)-I receptor (IGF-IR) mRNA and protein expression in both AR-positive (LNCaP cells) and AR-negative (PC-3 cells) prostate cancer cells. This effect occurs not only via ERα but also via ERβ stimulation and is specific for IGF-IR because it does not involve the cognate insulin receptor. IGF-IR up-regulation is associated with increased IGF-IR phosphorylation and with increased mitogenic and motogenic activities in response to IGF-I. IGF-IR up-regulation by E 2 does not require ER binding to DNA and is poorly sensitive to antiestrogen blockade, whereas it is associated with the activation of cytosolic kinase cascades involving Src, extracellular signal–regulated kinase (ERK)-1/2, and, to a lesser extent, phosphatidylinositol 3-kinase and is sensitive to the inhibition of these kinases. In conclusion, our data indicate that estrogens may contribute to IGF system deregulation in prostate cancer through the activation of a nongenotropic pathway. Estrogens may have a role, therefore, in tumor progression to androgen independence. Inhibition of the IGF-IR or the Src-ERK pathway should be considered, therefore, as an adjuvant therapy in prostate cancer. [Cancer Res 2007;67(18):8932–41]


PLOS ONE | 2012

Proline-rich tyrosine kinase 2 (Pyk2) regulates IGF-I-induced cell motility and invasion of urothelial carcinoma cells

Marco Genua; Shi-Qiong Xu; Simone Buraschi; Stephen C. Peiper; Leonard G. Gomella; Antonino Belfiore; Renato V. Iozzo; Andrea Morrione

The insulin-like growth factor receptor I (IGF-IR) plays an essential role in transformation by promoting cell growth and protecting cancer cells from apoptosis. We have recently demonstrated that the IGF-IR is overexpressed in invasive bladder cancer tissues and promotes motility and invasion of urothelial carcinoma cells. These effects require IGF-I-induced Akt- and MAPK-dependent activation of paxillin. The latter co-localizes with focal adhesion kinases (FAK) at dynamic focal adhesions and is critical for promoting motility of urothelial cancer cells. FAK and its homolog Proline-rich tyrosine kinase 2 (Pyk2) modulate paxillin activation; however, their role in regulating IGF-IR-dependent signaling and motility in bladder cancer has not been established. In this study we demonstrate that FAK was not required for IGF-IR-dependent signaling and motility of invasive urothelial carcinoma cells. On the contrary, Pyk2, which was strongly activated by IGF-I, was critical for IGF-IR-dependent motility and invasion and regulated IGF-I-dependent activation of the Akt and MAPK pathways. Using immunofluorescence and AQUA analysis we further discovered that Pyk2 was overexpressed in bladder cancer tissues as compared to normal tissue controls. Significantly, in urothelial carcinoma tissues there was increased Pyk2 localization in the nuclei as compared to normal tissue controls. These results provide the first evidence of a specific Pyk2 activity in regulating IGF-IR-dependent motility and invasion of bladder cancer cells suggesting that Pyk2 and the IGF-IR may play a critical role in the invasive phenotype in urothelial neoplasia. In addition, Pyk2 and the IGF-IR may serve as novel biomarkers with diagnostic and prognostic significance in bladder cancer.


Oncotarget | 2016

Suppression of progranulin expression inhibits bladder cancer growth and sensitizes cancer cells to cisplatin

Simone Buraschi; Shi Qiong Xu; Manuela Stefanello; Igor Moskalev; Alaide Morcavallo; Marco Genua; Ryuta Tanimoto; Ruth Birbe; Stephen C. Peiper; Leonard G. Gomella; Antonino Belfiore; Peter C. Black; Renato V. Iozzo; Andrea Morrione

We have recently demonstrated a critical role for progranulin in bladder cancer. Progranulin contributes, as an autocrine growth factor, to the transformed phenotype by modulating Akt-and MAPK-driven motility, invasion and anchorage-independent growth. Progranulin also induces F-actin remodeling by interacting with the F-actin binding protein drebrin. In addition, progranulin is overexpressed in invasive bladder cancer compared to normal tissue controls, suggesting that progranulin might play a key role in driving the transition to the invasive phenotype of urothelial cancer. However, it is not established whether targeting progranulin could have therapeutic effects on bladder cancer. In this study, we stably depleted urothelial cancer cells of endogenous progranulin by shRNA approaches and determined that progranulin depletion severely inhibited the ability of tumorigenic urothelial cancer cells to migrate, invade and grow in anchorage-independency. We further demonstrate that progranulin expression is critical for tumor growth in vivo, in both xenograft and orthotopic tumor models. Notably, progranulin levels correlated with response to cisplatin treatment and were upregulated in bladder tumors. Our data indicate that progranulin may constitute a novel target for therapeutic intervention in bladder tumors. In addition, progranulin may serve as a novel biomarker for bladder cancer.


Cancer Research | 2016

Abstract 698: Progranulin targeting in urothelial cancer cells inhibits motility, tumor growth in vitro and in vivo and sensitizes cells to cisplatin

Simone Buraschi; Shi-Qiong Xu; Manuela Stefanello; Igor Moskalev; Alaide Morcavallo; Marco Genua; Ryuta Tanimoto; Thomas Neill; Stephen C. Peiper; Leonard G. Gomella; Antonino Belfiore; Peter C. Black; Renato V. Iozzo; Andrea Morrione

Introduction and Objective: Bladder cancer is a major public health problem and affects more than 74,000 Americans with more than 16,000 estimated deaths in 2015. The majority of deaths are due to metastatic spread, commonly to the lungs. Understanding the molecular mechanisms regulating bladder tumor cell invasion and progression toward metastases is essential for developing better therapies to treat bladder cancer patients. The growth factor progranulin has emerged in recent years as an important regulator of transformation in several cancer models. We have previously established a critical role for progranulin in bladder cancer as in fact progranulin acts as an autocrine growth factor and promote motility and invasion of invasive urothelial cancer cells. In addition, progranulin is upregulated in high grade bladder cancer tissues compared to normal tissue controls suggesting that progranulin might work as a novel biomarker with predictive value for bladder cancer progression. However, whether progranulin is important for anchorage-independent growth and in vivo tumor formation of urothelial cancer cells has not been previously established. Methods: Progranulin depletion was achieved by stably transfecting tumorigenic T24T, UMUC3 urothelial cancer cells with a plasmid expressing an anti-progranulin shRNA. Progranulin-depleted and control UMUC-3 and T24T cells were tested for motility, invasion and anchorage-independent growth by soft-agar assays. Tumor formation in vivo was assessed in various UMUC-3-derived cell lines by xenograft and orthotopic models. Sensitivity to cisplatin was assessed by cell survival curves. Progranulin expression levels in a bladder tissue microarray were analyzed by HIC. Results: Progranulin-depleted T24T and UMUC-3 cells were significantly inhibited in their ability to migrate, close a wound and invade through Matrigel compared to control cells in both serum-deprived and 1% serum media. In addition, progranulin targeting strongly reduced the ability of T24T and UMUC-3 cells to grow in anchorage-independency and form colonies in soft-agar. Significantly progranulin-depleted UMUC-3 cells were severely inhibited in tumor formation in vivo as assessed by both xenograft and orthotopic models in immunocompromised mice. Importantly, progranulin depletion sensitized UMUC-3 cells to cisplatin. Finally, progranulin levels correlated with tumor progression in bladder cancer tissues. Conclusions: Our data are translationally relevant as indicate that progranulin exerts an essential functional role in the regulation of bladder cancer progression. Thus, progranulin may constitute a novel target for therapeutic intervention in bladder tumors. In addition, progranulin may serve as novel biomarker for diagnosis and prognosis of bladder cancer. Citation Format: Simone Buraschi, Shi-Qiong Xu, Manuela Stefanello, Igor Moskalev, Alaide Morcavallo, Marco Genua, Ryuta Tanimoto, Thomas Neill, Stephen C. Peiper, Leonard G. Gomella, Antonino Belfiore, Peter C. Black, Renato V. Iozzo, Andrea Morrione. Progranulin targeting in urothelial cancer cells inhibits motility, tumor growth in vitro and in vivo and sensitizes cells to cisplatin. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 698.


Cancer Research | 2015

Abstract 4945: A novel role for drebrin in regulating progranulin bioactivity in bladder cancer

Shi-Qiong Xu; Simone Buraschi; Alaide Morcavallo; Marco Genua; Tomoaki Shirao; Stephen C. Peiper; Leonard G. Gomella; Antonino Belfiore; Renato V. Iozzo; Andrea Morrione

Bladder cancer (BC) is one of the most common cancers in the United States with 56,390 estimated new cases and 11,170 estimated deaths in 2014. The majority of deaths is due to metastatic spread. Understanding the molecular mechanisms regulating bladder tumor cell invasion and progression toward metastases is essential for developing better therapies to treat bladder cancer patients. The growth factor progranulin plays an important role in cell proliferation, wound healing and transformation. We have recently established a critical role for progranulin in bladder cancer insofar as progranulin promotes urothelial cancer cell motility and contributes, as an autocrine growth factor, to the transformed phenotype by modulating invasion and anchorage-independent growth. In addition, progranulin expression is upregulated in invasive bladder cancer tissues compared to normal controls. However, the molecular mechanisms of progranulin action in bladder cancer have not been fully elucidated. In this study, we searched for novel progranulin-interacting proteins using pull-down assays with recombinant progranulin and proteomics. We discovered that drebrin, an F-actin binding protein, bound progranulin in urothelial cancer cells. We characterized drebrin function in various urothelial cancer cell lines and showed that drebrin is critical for progranulin-dependent activation of the Akt and MAPK pathways and modulates F-actin remodeling, motility, invasion and anchorage-independent growth. In addition, drebrin regulates tumor formation in vivo and its expression is upregulated in bladder cancer tissues compared to normal tissue controls. Our data are translationally relevant as indicate that drebrin exerts an essential functional role in the regulation of progranulin action and may constitute a novel target for therapeutic intervention in bladder tumors. In addition, drebrin may serve as novel biomarker for bladder cancer. This work was supported by the Benjamin Perkins Bladder Cancer Fund, National Institutes of Health Grants RO1 CA164462 (A.M., R.V.I.), and RO1 CA39481 and RO1 CA47282 (R.V.I.), Associazione Italiana per la Ricerca sul Cancro (AIRC) (grant n. 10625/12), AIRC project Calabria 2014 and Fondazione Cassa di Risparmio di Calabria e Lucania PON01_01078 (A.B.). Alaide Morcavallo was supported in part by Fondazione Diabete Ricerca. Citation Format: Shi-Qiong Xu, Simone Buraschi, Alaide Morcavallo, Marco Genua, Tomoaki Shirao, Stephen C. Peiper, Leonard G. Gomella, Antonino Belfiore, Renato V. Iozzo, Andrea Morrione. A novel role for drebrin in regulating progranulin bioactivity in bladder cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4945. doi:10.1158/1538-7445.AM2015-4945


Cancer Research | 2011

Abstract 1408: The insulin-like growth factor receptor I and Pyk2 kinase are up-regulated in urothelial carcinomas and promote motility and invasion of urothelial carcinoma-derived cells

Marco Genua; Shi-Qiong Xu; Leonard G. Gomella; Renato V. Iozzo; Andrea Morrione

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL The insulin-like growth factor receptor I (IGF-IR) plays an essential role in transformation by promoting cell growth and protecting cancer cells from apoptosis. We have recently demonstrated that the IGF-IR is overexpressed in invasive bladder cancer tissues and promotes motility and invasion of urothelial-carcinoma-derived cells. Significantly, IGF-IR activation did not affected cell proliferation, suggesting that the IGF-IR may play a more prominent role in promoting the transition to the invasive phenotype in bladder cancer. In this study we demonstrated by immunohistochemical analysis that the IGF-IR expression increased with grade in bladder cancer tissues compared to non-malignant controls. Increased IGF-IR expression in non-tumorigenic T24 urothelial carcinoma cells promoted anchorage independent growth, while targeting of endogenous IGF-IR by shRNA approaches in metastatic T24T cells inhibited colony formation in soft-agar. We also discovered that the focal adhesion protein Pyk2 kinase is activated by IGF-I in urothelial carcinoma-derived cells. Using siRNA approaches, we demonstrated that Pyk2 is critical for IGF-IR-dependent motility and invasion of urothelial cancer cells. In addition, Pyk 2 is critical for IGF-I-dependent activation of the Akt and MAPK pathways, which are critical in the regulation of motility and invasion. Using immunofluorescence and AQUA (Automated Quantitative Analysis) analysis we also discovered that Pyk2 is overexpressed in bladder cancer tissues compared to normal tissue controls. In urothelial carcinoma-derived tissues there is also increased Pyk2 localization in the nucleus compared to normal tissue controls. Our results provide the first evidence for a role of the IGF-IR and Pyk2 in motility and invasion of bladder cancer cells, and support the hypothesis that the IGF-IR and Pyk2 may play a critical role in the establishment of the invasive phenotype in urothelial neoplasia. The IGF-IR and Pyk2 could represent novel molecular targets in bladder cancer and could also serve as a novel tumor biomarkers for diagnosis and possibly prognosis of bladder tumors. This work has been supported by the Benjamin Perkins Bladder Cancer Fund, the Martin Greitzer Fund and National Institutes of Health Grants RO1 DK068419 (A.M.) and RO1 CA39481 and RO1 CA047282 (R.V.I.). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1408. doi:10.1158/1538-7445.AM2011-1408


Cancer Research | 2009

Role of Cyclic AMP Response Element–Binding Protein in Insulin-like Growth Factor-I Receptor Up-regulation by Sex Steroids in Prostate Cancer Cells

Marco Genua; Giuseppe Pandini; Diego Sisci; Gabriella Castoria; Marcello Maggiolini; Riccardo Vigneri; Antonino Belfiore

Collaboration


Dive into the Marco Genua's collaboration.

Top Co-Authors

Avatar

Andrea Morrione

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Renato V. Iozzo

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Leonard G. Gomella

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shi-Qiong Xu

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Simone Buraschi

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Stephen C. Peiper

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Alaide Morcavallo

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Manuela Stefanello

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge