Network


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

Hotspot


Dive into the research topics where Saverio Bettuzzi is active.

Publication


Featured researches published by Saverio Bettuzzi.


International Journal of Cancer | 2004

Clusterin (SGP‐2, ApoJ) expression is downregulated in low‐ and high‐grade human prostate cancer

Maurizio Scaltriti; Maurizio Brausi; Andrea Amorosi; Andrea Caporali; Domenico D'Arca; Serenella Astancolle; Arnaldo Corti; Saverio Bettuzzi

Clusterin is overexpressed during tissue and cell involution and downregulated in proliferating cells. Its role in cell survival, cell death and neoplastic transformation remains debated. We studied the expression and distribution of clusterin mRNA and protein in human prostate carcinoma (CaP) specimens of different degrees of malignancy. Fresh CaP specimens were obtained from 25 patients subjected to long‐term androgen ablation before surgery. Clusterin expression was studied by Northern and Western analysis, in situ hybridization and immunohistochemistry, in comparison with Gas1 and histone H3 mRNA (markers of cell quiescence and S phase of the cell cycle, respectively). Clusterin is downregulated in CaP in comparison with matched benign controls. In low‐grade CaP, clusterin colocalized with Gas1 to the stromal compartment, and in some glands, the basal lamina was heavily stained. In high‐grade CaP clusterin stained the remnants of stromal matrix while histone H3 localized to cancer cells, which were very rarely clusterin positive. High clusterin expression was found in the branches of a nerve infiltrated by tumor. The periglandular clusterin expression found in low‐grade CaP could result from induction of quiescence and/or apoptosis of prostatic fibroblasts lining those glands in which tumor invasion is at an initial stage, involving basal lamina. In advanced CaP, the staining of the remnants of the extracellular matrix suggests a role for clusterin in the process of dismantling the stromal organization caused by cancer progression.


Cancer Research | 2004

Intracellular Clusterin Induces G2-M Phase Arrest and Cell Death in PC-3 Prostate Cancer Cells1

Maurizio Scaltriti; Anna Santamaria; Rosanna Paciucci; Saverio Bettuzzi

Enhanced clusterin gene expression has been related frequently to organ remodeling, tissue involution, and cell death. Whether clusterin represents a leading cause or a consequence of apoptosis induction is still a matter of debate. Clusterin is known as an extracellular secreted glycoprotein in the mature form. However, truncated isoforms of the protein and nuclear localization of clusterin have been described recently in association to cell death. Here, we show the biological effects triggered in PC-3 androgen-independent prostate cancer cells by overexpression of an intracellular, not secreted form of clusterin (intracellular-clusterin). Transient transfection of PC-3 cells with intracellular-clusterin resulted in nuclear localization signal-independent massive nuclear localization of the protein leading to G2-M phase blockade followed by caspase-dependent apoptosis. Constitutive expression of intracellular-clusterin (pFLAG- intracellular-clusterin) in recombinant PC-3 cells caused clonogenic toxicity. The rare pFLAG-intracellular clusterin surviving clones showed inhibition of the proliferation rate and altered phenotype with impaired mitosis and endoreduplication. In these cells, caspase-independent cell death was induced. Impaired cell cycle progression in pFLAG-intracellular-clusterin clones was associated to arrest at the G2-M checkpoint by down-regulation of the mitotic complex cyclin B1/cyclin-dependent kinase 1. Intriguingly, intracellular-clusterin was localized exclusively in the cytoplasm in stably transfected cells, suggesting a negative correlation between nuclear clusterin accumulation and cell survival. These findings may possibly explain the conflicting results obtained in different laboratories, suggesting that clusterin might be a proapoptotic or a survival gene, also opening new perspectives for the characterization of androgen-independent and apoptosis-resistant prostate cancer cells.


Cancer Research | 2004

Clusterin-Mediated Apoptosis Is Regulated by Adenomatous Polyposis Coli and Is p21 Dependent but p53 Independent

Tingan Chen; Joel G. Turner; Susan McCarthy; Maurizio Scaltriti; Saverio Bettuzzi; Timothy J. Yeatman

Clusterin is a widely expressed glycoprotein that has been paradoxically observed to have both pro- and antiapoptotic functions. Recent reports suggest this apparent dichotomy of function may be related to two different isoforms, one secreted and cytoplasmic, the other nuclear. To clarify the functional role of clusterin in regulating apoptosis, we examined its expression in human colon cancer tissues and in human colon cancer cell lines. We additionally explored its expression and activity using models of adenomatous polyposis coli (APC)- and chemotherapy-induced apoptosis. Clusterin RNA and protein levels were decreased in colon cancer tissues largely devoid of wild-type APC when compared with matched normal tissue controls, suggesting a means for invasive cancers to avoid apoptosis. Conversely, induction of apoptosis by expression of wild-type APC or by treatment with chemotherapy led to increased clusterin RNA and protein levels localizing to apoptotic nuclei. We found that transient transfection of clusterin to colon cancer cell lines directly enhanced basal and chemotherapy-induced apoptosis. Clusterin-induced apoptosis was inhibited by antisense clusterin and was found to be highly dependent on p21 but not p53 expression, yet a deficit in p21 can be subverted by clusterin transfection. Collectively, these data support the hypothesis that nuclear clusterin function is proapoptotic when induced by APC or chemotherapy in the context of p21 expression. Absent of p21, clusterin in not induced, and apoptosis is significantly inhibited. These data support a potential therapeutic role for clusterin in enhancing chemotherapy-induced apoptosis and in promoting apoptosis in cells deficient in p21.


Oncogene | 2002

Clusterin (SGP-2) transient overexpression decreases proliferation rate of SV40-immortalized human prostate epithelial cells by slowing down cell cycle progression

Saverio Bettuzzi; Francesca Scorcioni; Serenella Astancolle; Pierpaola Davalli; Maurizio Scaltriti; Arnaldo Corti

Clusterin is a highly conserved, widely distributed glycoprotein whose biological significance is still debated. Involved in many biological processes and disease states, clusterin is induced by cell injury and tissue regression, but is repressed during cell proliferation. We have previously reported that clusterin mRNA induction is associated with epithelial cell atrophy in the rat prostate and both clusterin transcript and protein accumulated in quiescent normal human skin fibroblasts. Here we show that transient clusterin overexpression, in SV40-immortalized human prostate epithelial cells (PNT2), resulted in increased accumulation of cells in the G0/G1 phases of the cell cycle, accompanied by slowdown of cell cycle progression and decrease of DNA synthesis. The activities of ornithine decarboxylase (ODC) and S-adenosylmethionine decarboxylase (AdoMetDC), and the level of histone H3 mRNA (markers of cell proliferation) concomitantly decreased, while Gas1 mRNA (a marker of cell quiescence) accumulated. Thus it appears that clusterin, by opposing the effect of SV40 on the proliferation rate of PNT2 cells, acts as an anti-oncogene in the prostate, suggesting a role for this gene in controlling proliferation of normal and transformed prostate epithelial cells.


Cancer and Metastasis Reviews | 2008

Demethylation of (Cytosine-5-C-methyl) DNA and regulation of transcription in the epigenetic pathways of cancer development

Samir Kumar Patra; Aditi Patra; Federica Rizzi; Tapash Chandra Ghosh; Saverio Bettuzzi

Cancer cells and tissues exhibit genome wide hypomethylation and regional hypermethylation. CpG-methylation of DNA (MeCpG-DNA) is defined as the formation of a C–C covalent bond between the 5′-C of cytosine and the –CH3 group of S-adenosylmethionine. Removal of the sole –CH3 group from the methylated cytosine of DNA is one of the many ways of DNA-demethylation, which contributes to activation of transcription. The mechanism of demethylation, the candidate enzyme(s) exhibiting direct demethylase activity and associated cofactors are not firmly established. Genome-wide hypomethylation can be obtained in several ways by inactivation of DNMT enzyme activity, including covalent trapping of DNMT by cytosine base analogues. Removal of methyl layer could also be occurred by excision of the 5-methyl cytosine base by DNA glycosylases. The importance of truly chemically defined direct demethylation of intact DNA in regulation of gene expression, development, cell differentiation and transformation are discussed in this contribution.


Endocrine-related Cancer | 2010

The clusterin paradigm in prostate and breast carcinogenesis

Federica Rizzi; Saverio Bettuzzi

The biological functions of clusterin (CLU, also known as ApoJ, SGP2, TRPM-2, CLI) have been puzzling the researchers since its first discovery in the early 80s. We know that CLU is a single 9-exons gene expressing three protein forms with different sub-cellular localisations and diverse biological functions. Despite the many reports from many research teams on CLU action and its relation to tumourigenesis, contradictions in the data and alternative hypothesis still exist. Understanding the role of CLU in tumourigenesis is complicated not only by the existence of different protein forms but also by the changes of tumours over time and the selection pressures imposed by treatments such as hormone ablation or chemotherapy. This review focuses on recent discoveries concerning the role of CLU in prostate and breast cancer onset and progression. Although CLU acts primarily as a tumour suppressor in the early stages of carcinogenesis, consistent with its role in the involution of the prostate following castration, late stage cancer may overexpress CLU following chemotherapeutic drugs or hormonal ablation therapy. High expression of secreted or cytoplasmic CLU may represent a pro-survival stimulus because it confers increased resistance to killing by anti-cancer drugs or enhances tumour cell survival in specific niches.


Journal of the National Cancer Institute | 2009

Clusterin, a haploinsufficient tumor suppressor gene in neuroblastomas.

Olesya Chayka; Daisy Corvetta; Michael Dews; Alessandro E. Caccamo; Izabela Piotrowska; Giorgia Santilli; Sian Gibson; Nj Sebire; Nourredine Himoudi; Michael D. Hogarty; John Anderson; Saverio Bettuzzi; Andrei Thomas-Tikhonenko; Arturo Sala

BACKGROUND Clusterin expression in various types of human cancers may be higher or lower than in normal tissue, and clusterin may promote or inhibit apoptosis, cell motility, and inflammation. We investigated the role of clusterin in tumor development in mouse models of neuroblastoma. METHODS We assessed expression of microRNAs in the miR-17-92 cluster by real-time reverse transcription-polymerase chain reaction in MYCN-transfected SH-SY5Y and SH-EP cells and inhibited expression by transfection with microRNA antisense oligonucleotides. Tumor development was studied in mice (n = 66) that were heterozygous or homozygous for the MYCN transgene and/or for the clusterin gene; these mice were from a cross between MYCN-transgenic mice, which develop neuroblastoma, and clusterin-knockout mice. Tumor growth and metastasis were studied in immunodeficient mice that were injected with human neuroblastoma cells that had enhanced (by clusterin transfection, four mice per group) or reduced (by clusterin short hairpin RNA [shRNA] transfection, eight mice per group) clusterin expression. All statistical tests were two-sided. RESULTS Clusterin expression increased when expression of MYCN-induced miR-17-92 microRNA cluster in SH-SY5Y neuroblastoma cells was inhibited by transfection with antisense oligonucleotides compared with scrambled oligonucleotides. Statistically significantly more neuroblastoma-bearing MYCN-transgenic mice were found in groups with zero or one clusterin allele than in those with two clusterin alleles (eg, 12 tumor-bearing mice in the zero-allele group vs three in the two-allele group, n = 22 mice per group; relative risk for neuroblastoma development = 4.85, 95% confidence interval [CI] = 1.69 to 14.00; P = .005). Five weeks after injection, fewer clusterin-overexpressing LA-N-5 human neuroblastoma cells than control cells were found in mouse liver or bone marrow, but statistically significantly more clusterin shRNA-transfected HTLA230 cells (3.27%, with decreased clusterin expression) than control-transfected cells (1.53%) were found in the bone marrow (difference = 1.74%, 95% CI = 0.24% to 3.24%, P = .026). CONCLUSIONS We report, to our knowledge, the first genetic evidence that clusterin is a tumor and metastasis suppressor gene.


Genomics | 1991

The gene for SP-40,40 human homolog of rat sulfated glycoprotein 2, rat clusterin, and rat testosterone-repressed prostate message 2, maps to chromosome 8

Michele Purrello; Saverio Bettuzzi; Cinzia Di Pietro; Elena Mirabile; Martina Di Blasi; Rebecca Rimini; Karl-Heinz Grzeschik; Cristina Ingletti; Arnaldo Corti; Giovanni Sichel

Sulfated glycoprotein 2 (SGP-2) is a rat glycoprotein that is particularly abundant in seminal fluid, where it is found associated with the acrosome and the tail of mature spermatozoa; for this reason it has been suggested that it has an important role in spermatogenesis. On the basis of nucleotide sequence homology, it has been proposed that the orthologous human gene is that coding for serum protein-40,40 (SP-40,40), a serum protein also called complement lysis inhibitor (CLI), SP-40,40 has been shown to act as a control mechanism of the complement cascade: in fact, it prevents the binding of a C5b-C7 complex to the membrane of the target cell and in this way inhibits complement-mediated cytolysis. SGP-2 and SP-40,40 seem then to be part of different biological systems. Furthermore it has been shown that another protein, testosterone-repressed prostate message 2 (TRPM-2), shares sequence homology with SGP-2 and SP-40,40. TRPM-2 is expressed at high levels and in a temporally precisely defined manner in dying cells, an observation that would suggest its involvement in the cascade of events leading to cell death. We have used a large panel of 24 mouse/human hybrid cell lines and a cDNA for SGP-2, which is also highly homologous to that for rat clusterin, to map the chromosomal location of the orthologous human gene. The mapping data and the Southern analysis presented in this paper, in addition to the data available from the literature, strongly suggest that in the human genome there is a single locus homologous to the probe used and that it codes for the protein which has been called, in different species, SP-40,40, SGP-2, clusterin, and TRPM-2. The chromosomal mapping of the locus for this multiname protein should facilitate its cloning and a better understanding of the apparently many biological functions of its product.


Biochemical Journal | 2004

Cell detachment and apoptosis induction of immortalized human prostate epithelial cells are associated with early accumulation of a 45 kDa nuclear isoform of clusterin

Alessandro E. Caccamo; Maurizio Scaltriti; Andrea Caporali; Domenico D'Arca; Francesca Scorcioni; Serenella Astancolle; Massimo Mangiola; Saverio Bettuzzi

Clusterin, ubiquitously distributed in mammalians, was cloned and identified as the most potently induced gene during rat prostate involution following androgen deprivation. Also found to be involved in many other patho-physiological processes, its biological significance is still controversial, particularly with regard to apoptosis. We previously showed that transient over-expression of clusterin blocked cell cycle progression of simian-virus-40-immortalized human prostate epithelial cell lines PNT1A and PNT2. We show in the present study that the accumulation of an intracellular 45 kDa clusterin isoform was an early event closely associated with death of PNT1A cells caused by cell detachment followed by apoptosis induction (anoikis). Cell morphological changes, decreased proliferation rate and cell cycle arrest at G0/G1-S-phase checkpoint were all strictly associated with the production and early translocation to the nucleus of a 45 kDa clusterin isoform. Later, nuclear clusterin was found accumulated in detached cells and apoptotic bodies. These results suggest that a 45 kDa isoform of clusterin, when targeted to the nucleus, can decrease cell proliferation and promotes cell-detachment-induced apoptosis, suggesting a possible major role for clusterin as an anti-proliferative gene in human prostate epithelial cells.


Cell Death & Differentiation | 2005

Ca2+ depletion induces nuclear clusterin, a novel effector of apoptosis in immortalized human prostate cells

Alessandro E. Caccamo; Maurizio Scaltriti; Andrea Caporali; Domenico D'Arca; Arnaldo Corti; D Corvetta; A Sala; Saverio Bettuzzi

Ca 2+ depletion induces nuclear clusterin, a novel effector of apoptosis in immortalized human prostate cells

Collaboration


Dive into the Saverio Bettuzzi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arnaldo Corti

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Serenella Astancolle

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Maurizio Scaltriti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pierpaola Davalli

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Domenico D'Arca

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge