Network


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

Hotspot


Dive into the research topics where Maria Vincenza Chiantore is active.

Publication


Featured researches published by Maria Vincenza Chiantore.


International Journal of Cancer | 2013

Successful therapeutic vaccination with integrase defective lentiviral vector expressing nononcogenic human papillomavirus E7 protein.

Felicia Grasso; Donatella R.M. Negri; Stefania Mochi; A. R. Rossi; Armando Cesolini; Andrea Giovannelli; Maria Vincenza Chiantore; Pasqualina Leone; Colomba Giorgi; Andrea Cara

Persistent infection with high risk genotypes of human papillomavirus (HPV) is the cause of cervical cancer, one of most common cancer among woman worldwide, and represents an important risk factor associated with other anogenital and oropharyngeal cancers in men and women. Here, we designed a therapeutic vaccine based on integrase defective lentiviral vector (IDLV) to deliver a mutated nononcogenic form of HPV16 E7 protein, considered as a tumor specific antigen for immunotherapy of HPV‐associated cervical cancer, fused to calreticulin (CRT), a protein able to enhance major histocompatibility complex class I antigen presentation (IDLV‐CRT/E7). Vaccination with IDLV‐CRT/E7 induced a potent and persistent E7‐specific T cell response up to 1 year after a single immunization. Importantly, a single immunization with IDLV‐CRT/E7 was able to prevent growth of E7‐expressing TC‐1 tumor cells and to eradicate established tumors in mice. The strong therapeutic effect induced by the IDLV‐based vaccine in this preclinical model suggests that this strategy may be further exploited as a safe and attractive anticancer immunotherapeutic vaccine in humans.


PLOS ONE | 2012

Interferon-β Induces Cellular Senescence in Cutaneous Human Papilloma Virus-Transformed Human Keratinocytes by Affecting p53 Transactivating Activity

Maria Vincenza Chiantore; Serena Vannucchi; Rosita Accardi; Massimo Tommasino; Zulema A. Percario; Gabriele Vaccari; Elisabetta Affabris; Gianna Fiorucci; Giovanna Romeo

Interferon (IFN)-β inhibits cell proliferation and affects cell cycle in keratinocytes transformed by both mucosal high risk Human Papilloma Virus (HPV) and cutaneous HPV E6 and E7 proteins. In particular, upon longer IFN-β treatments, cutaneous HPV38 expressing cells undergo senescence. IFN-β appears to induce senescence by upregulating the expression of the tumor suppressor PML, a well known IFN-induced gene. Indeed, experiments in gene silencing via specific siRNAs have shown that PML is essential in the execution of the senescence programme and that both p53 and p21 pathways are involved. IFN-β treatment leads to a modulation of p53 phosphorylation and acetylation status and a reduction in the expression of the p53 dominant negative ΔNp73. These effects allow the recovery of p53 transactivating activity of target genes involved in the control of cell proliferation. Taken together, these studies suggest that signaling through the IFN pathway might play an important role in cellular senescence. This additional understanding of IFN antitumor action and mechanisms influencing tumor responsiveness or resistance appears useful in aiding further promising development of biomolecular strategies in the IFN therapy of cancer.


Cancer Chemotherapy and Pharmacology | 2000

Differences in uptake and metabolism of retinoic acid between estrogen receptor-positive and -negative human breast cancer cells.

Kosuke Okamoto; Fausto Andreola; Maria Vincenza Chiantore; Robert L. Dedrick; Luigi M. De Luca

Purpose: Our previous work had shown that retinoic acid (RA) inhibits cell growth and induces apoptosis in estrogen receptor-positive (ER-positive) MCF-7 and T-47D human breast carcinoma cells, but not in ER-negative human breast carcinoma cells MB-231 and MB-453. The purpose of this work was to determine whether these differences might be due to differences in uptake and metabolism of the drug between ER-positive and ER-negative cells. Methods: We measured RA uptake in cultured human breast cancer cells and determined its metabolism by high-pressure liquid chromatographic analysis. Results: The two ER-positive cell lines reached maximum RA uptake at about 2u2009h, followed by a sharp decline, so that most RA had disappeared from the cells and from the medium by 24u2009h and was found as oxidation products in the culture medium. In contrast, the two ER-negative cell lines showed a pattern of lower accumulation without the sharp increase and subsequent steep decline, so that by 24u2009h there was more RA in these cells and their culture medium than in the RA-responsive ER-positive cells, even though at 2u2009h the ER-negative cells had taken up less RA than the ER-positive cells. Kinetic analysis of the uptake of RA in MCF-7 cells was consistent with rapid movement across the cell membranes and the actual rate determined by diffusion of albumin-bound retinoid to the cells. Conclusions: This study is the first to demonstrate profound differences in RA accumulation and confirms previous results on different rates of RA metabolism between ER-positive and ER-negative human breast cancer cells. The findings reported here, therefore, may introduce additional elements to be considered in the design of new drugs for cancer chemoprevention and therapy.


Journal of Cancer Research and Clinical Oncology | 2016

Human papillomavirus E6 and E7 oncoproteins affect the expression of cancer-related microRNAs: additional evidence in HPV-induced tumorigenesis

Maria Vincenza Chiantore; Giorgio Mangino; Marco Iuliano; Maria Simona Zangrillo; Ilaria De Lillis; Gabriele Vaccari; Rosita Accardi; Massimo Tommasino; Sandra Columba Cabezas; Maurizio Federico; Gianna Fiorucci; Giovanna Romeo

PurposeHuman papillomaviruses (HPVs) are the causative agents of cervical cancer and are also associated with other types of cancers. HPVs can modulate microRNAs (miRNAs) expressed by infected cells. The production of extracellular vesicles is deregulated in cancer, and their cargo delivered to the microenvironment can promote tumorigenesis. The involvement of HPV oncoproteins on miRNA expression in cells and exosomes was analyzed in keratinocytes transduced with E6 and E7 from mucosal HPV-16 or cutaneous HPV-38 (K16 and K38).MethodsMiRNAs were investigated through the TaqMan Array Human MicroRNA Cards, followed by real-time RT-PCR assay for specific miRNAs. Selected miRNA targets were analyzed by Western blot and correlated to the HPV oncoproteins by specifically silencing E6 and E7 expression. Exosomes, isolated from K16 and K38 supernatants by differential centrifugations, were quantified through the vesicle-associated acetylcholinesterase activity.ResultsMiRNAs deregulated in K16 and K38 cells were identified. HPV-16 and/or HPV-38 E6 and E7 single proteins can modify the expression of selected miRNAs involved in the tumorigenesis, in particular miR-18a, -19a, -34a and -590-5p. The analysis of the content of exosomes isolated from HPV-positive cells revealed the presence of E6 and E7 mRNAs and few miRNAs. MiR-222, a key miRNA deregulated in many cancers, was identified in exosomes from K16 cells.ConclusionsHPV E6 and/or E7 oncoprotein expression can induce the deregulation of some miRNAs. Through the production and function of exosomes, HPV oncogenes as well as HPV-deregulated miRNAs can potentiate the virus oncogenic effects in the tumor cell microenvironment.


Retrovirology | 2012

Simian immunodeficiency virus-Vpx for improving integrase defective lentiviral vector-based vaccines

Donatella R.M. Negri; A. R. Rossi; Maria Blasi; Zuleika Michelini; Pasqualina Leone; Maria Vincenza Chiantore; Silvia Baroncelli; Gemma Perretta; Andrea Cimarelli; Mary E. Klotman; Andrea Cara

BackgroundIntegrase defective lentiviral vectors (IDLV) represent a promising delivery system for immunization purposes. Human dendritic cells (DC) are the main cell types mediating the immune response and are readily transduced by IDLV, allowing effective triggering of in vitro expansion of antigen-specific primed CD8+ T cells. However, IDLV expression in transduced DC is at lower levels than those of the integrase (IN) competent counterpart, thus requiring further improvement of IDLV for future use in the clinic.ResultsIn this paper we show that the addition of simian immunodeficiency (SIV)-Vpx protein in the vector preparation greatly improves transduction of human and simian DC, but not of murine DC, thus increasing the ability of transduced DC to act as functional antigen presenting cells, in the absence of integrated vector sequences. Importantly, the presence of SIV-Vpx allows for using lower dose of input IDLV during in vitro transduction, thus further improving the IDLV safety profile.ConclusionsThese results have significant implications for the development of IDLV-based vaccines.


Cytokine & Growth Factor Reviews | 2016

Inflammatory microenvironment and human papillomavirus-induced carcinogenesis

Giorgio Mangino; Maria Vincenza Chiantore; Marco Iuliano; Gianna Fiorucci; Giovanna Romeo

More than 15% of the global cancer burden is attributable to infectious agents. Pathogens that cause persistent infections are strongly associated with cancer, inflammation being a major component of the chronic infections as revealed by basic, clinical and epidemiological studies. Persistent infection and viral oncoproteins induce specific cellular pathways modifications that promote tumorigenesis. Deregulated and continuous immune response leads to severe tissue and systemic damage, impaired tumor surveillance and consequent carcinogenesis promotion by selecting for metastatic and therapeutically resistant tumor phenotypes. In this review, the role of inflammatory microenvironment in the HPV-induced carcinogenesis is addressed, with a specific focus on the involvement of the immune molecules and microRNAs as well as their delivery through the microvesicle cargo.


Cytokine & Growth Factor Reviews | 2015

MicroRNAs in virus-induced tumorigenesis and IFN system

Gianna Fiorucci; Maria Vincenza Chiantore; Giorgio Mangino; Giovanna Romeo

Numerous microRNAs (miRNAs), small non-coding RNAs encoded in the human genome, have been shown to be involved in cancer pathogenesis and progression. There is evidence that some of these miRNAs possess proapoptotic or proliferation promoting roles in the cell by negatively regulating target mRNAs. Oncogenic viruses are able to produce persistent infection, favoring tumor development by deregulating cell proliferation and inhibiting apoptosis. It has been recently suggested that cellular miRNAs may participate in host-virus interactions, influencing viral replication. Many mammalian viruses counteract this cellular antiviral defense by using viral proteins but also by encoding viral miRNAs involved in virus-induced tumorigenesis. Interferons (IFNs) modulate a number of non-coding RNA genes, especially miRNAs, that may be used by mammalian organisms as a mechanism of IFN system to combat viral infection and related diseases. In particular, IFNs might induce specific cellular miRNAs that target viral transcripts thereby using this strategy as part of their effectiveness against invading viruses. Therefore IFNs, interferon stimulated genes and miRNAs could act synergistically as innate response to virus infection to induce a potent non-permissive cellular environment for virus replication and virus-induced cancer. The relevance of this reviewed research topic is clearly related to the observation that although virus infections are responsible of specific tumors, other unidentified genetic alterations are likely involved in the induction of malignant transformation. The identification of such genetic alterations, i.e. miRNA expression in transformed cells, would be of considerable importance for the analysis of the pathogenesis and for the treatment of cancer induced by specific viruses as well as for the advancement of the current knowledge on the molecular mechanisms underlying virus-host interaction. In this respect, we will review also the important, still little explored, roles of miRNAs acting both as IFN-stimulated anti-viral molecules and as critical regulators of IFNs and IFN-stimulated genes.


Cytokine | 2017

IFN-β antiproliferative effect and miRNA regulation in Human Papilloma Virus E6- and E7-transformed keratinocytes

Maria Vincenza Chiantore; Giorgio Mangino; Marco Iuliano; Maria Simona Zangrillo; Ilaria De Lillis; Gabriele Vaccari; Rosita Accardi; Massimo Tommasino; Gianna Fiorucci; Giovanna Romeo

HighlightsHPV E6 and E7 oncoproteins are involved in IFN‐&bgr; antiproliferative effects.E6‐dependent p53 inhibition is correlated with senescence resistance in keratinocytes.IFN modulates the expression of selected microRNAs depending on the cellular context.MiRNA regulation is an important mechanism of the IFN‐&bgr; antiproliferative effects. ABSTRACT Human Papilloma Viruses (HPVs) are the causative agents of cervical cancer although other types of cancers are associated with HPV infection. Type I Interferons can interfere with HPV E6‐ and/or E7‐dependent transformation and can affect microRNA (miRNA) expression. Cancer cells show a specific pattern of miRNA expression and HPVs are able to modulate miRNAs expressed in infected cells. Keratinocytes transduced with E6 and E7 from mucosal HPV‐16 or cutaneous HPV‐38 (K16 and K38) were studied to analyze the involvement of HPV oncoproteins in the anti‐proliferative activity of IFN‐&bgr;. In view of our previous data showing senescence induction by the cytokine in K38 cells, we observe that IFN‐&bgr; treatment leads to p53‐indipendent apoptosis in K16 cells whereas induces senescence in K16 cells if E6 is silenced and p53 expression is restored. The levels of selected miRNAs, deregulated in K16 and K38 cells, can be modulated by IFN‐&bgr; when E6 and E7 proteins of HPV‐16, but not HPV‐38, are expressed.


Cytokine | 2017

Human Papillomavirus E6 and E7 oncoproteins affect the cell microenvironment by classical secretion and extracellular vesicles delivery of inflammatory mediators

Marco Iuliano; Giorgio Mangino; Maria Vincenza Chiantore; Maria Simona Zangrillo; Rosita Accardi; Massimo Tommasino; Gianna Fiorucci; Giovanna Romeo

HighlightsHPV E6 and E7 oncoproteins affect the inflammatory mediators expression.HPV E6 and E7 silencing demonstrates the specificity of the inflammatory mediators modulation.Inflammatory cytokines and chemokines are secreted in the extracellular milieu.Extracellular vesicles released by HPV‐positive cells deliver a modified cargo of inflammatory mediators. &NA; The connection between chronic inflammation and risk of cancer has been supported by several studies. The development of cancer might be a process driven by the presence of a specific combination of inflammatory mediators, including cytokines, chemokines and enzymes, in the tumor microenvironment. Virus‐induced tumors, like HPV‐induced Squamous Cell Carcinomas, represent a paradigmatic example of the interplay between inflammation, as integral part of the innate antiviral response, and malignant transformation. Here, the role of inflammatory microenvironment in the HPV‐induced carcinogenesis is addressed, with a specific focus on the involvement of the immune molecules as well as their delivery through the microvesicle cargo possibly correlated to the different HPV genotype. The expression of the inflammatory mediators in HPV positive cells has been analyzed in primary human foreskin keratinocytes and keratinocytes transduced by E6 and E7 from mucosal HPV‐16 or cutaneous HPV‐38 genotypes. HPV E6 and E7 proteins can modulate the expression of immune mediators in HPV‐infected cells and can affect the levels of immune molecules, mainly chemokines, in the extracellular milieu. HPV‐16 E6 and E7 oncoproteins have been silenced to confirm the specificity of the modulation of the inflammatory microenvironment. Our results suggest that the expression of HPV oncoproteins allows the modification of the tumor milieu through the synthesis and release of specific pro‐inflammatory cytokines and chemokines, affecting the efficacy of the immune response. The microenvironment can also be conditioned by an altered mRNA cargo delivered by extracellular vesicles, thereby efficiently affecting the surrounding cells with possible implication for tumorigenesis and tumor diagnosis.


Biochemical and Biophysical Research Communications | 1997

TPA Induces Transglutaminase C and Inhibits Cell Growth in the Colon Carcinoma Cell Line SW620

Karolina Kósa; M.I. Rosenberg; Maria Vincenza Chiantore; L. M. De Luca

Collaboration


Dive into the Maria Vincenza Chiantore's collaboration.

Top Co-Authors

Avatar

Gianna Fiorucci

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Giovanna Romeo

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Massimo Tommasino

International Agency for Research on Cancer

View shared research outputs
Top Co-Authors

Avatar

Rosita Accardi

International Agency for Research on Cancer

View shared research outputs
Top Co-Authors

Avatar

Giorgio Mangino

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Marco Iuliano

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Serena Vannucchi

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Gabriele Vaccari

Istituto Superiore di Sanità

View shared research outputs
Researchain Logo
Decentralizing Knowledge