Network


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

Hotspot


Dive into the research topics where Vincenzo Ciminale is active.

Publication


Featured researches published by Vincenzo Ciminale.


Journal of Virology | 2001

Free Major Histocompatibility Complex Class I Heavy Chain Is Preferentially Targeted for Degradation by Human T-Cell Leukemia/Lymphotropic Virus Type 1 p12I Protein

Julie M. Johnson; Christophe Nicot; Jake Fullen; Vincenzo Ciminale; Luca Casareto; James C. Mulloy; Steve Jacobson; Genoveffa Franchini

ABSTRACT Human T-cell leukemia virus type 1 (HTLV-1) establishes a persistent infection in the host despite a vigorous virus-specific immune response. Here we demonstrate that an HTLV-1-encoded protein, p12I, resides in the endoplasmic reticulum (ER) and Golgi and physically binds to the free human major histocompatibility complex class I heavy chains (MHC-I-Hc) encoded by the HLA-A2, -B7, and -Cw4 alleles. As a result of this interaction, the newly synthesized MHC-I-Hc fails to associate with β2-microglobulin and is retrotranslocated to the cytosol, where it is degraded by the proteasome complex. Targeting of the free MHC-I-Hc, and not the MHC-I-Hc–β2-microglobulin complex, by p12Irepresents a novel mechanism of viral interference and disrupts the intracellular trafficking of MHC-I, which results in a significant decrease in surface levels of MHC-I on human T-cells. These findings suggest that the interaction of p12I with MHC-1-Hc may interfere with antigen presentation in vivo and facilitate escape of HTLV-1-infected cells from immune recognition.


Oncogene | 2005

Human T-cell leukemia/lymphoma virus type 1 nonstructural genes and their functions

Christophe Nicot; Robert Harrod; Vincenzo Ciminale; Genoveffa Franchini

The human T-cell leukemia/lymphoma virus (HTLV) genome, in addition to the structural Gag and Env proteins and retroviral enzymes, carries a region at its 3′ end originally designated pX. To date, we know that this region encodes two essential transcriptional and post-transcriptional positive regulators of viral expression, the Tax and Rex proteins, respectively (reviewed elsewhere in this issue). Here, we will review current knowledge of the functions of three additional proteins encoded in the pX region, p12I, p13II, and p30II.


Oncogene | 1999

Mitochondrial targeting of the p13II protein coded by the x-II ORF of human T-cell leukemia/lymphotropic virus type I (HTLV-I).

Vincenzo Ciminale; Lorenza Zotti; Donna M. D'Agostino; Tiziana Ferro; Luca Casareto; Genoveffa Franchini; Paolo Bernardi; Luigi Chieco-Bianchi

The X region of the HTLV-I genome contains four major open reading frames (ORFs), two of which, termed x-I and x-II, are of still undefined biological significance. By indirect immunofluorescence and dual labeling with marker proteins, we demonstrate that p13II, an 87-amino acid protein coded by the x-II ORF, is selectively targeted to mitochondria. Mutational analysis revealed that mitochondrial targeting of p13II is directed by an atypical 10-amino acid signal sequence that is not cleaved upon import and is able to target the Green Fluorescent Protein to mitochondria. Expression of p13II results in specific alterations of mitochondrial morphology and distribution from a typical string-like, dispersed network to round-shaped clusters, suggesting that p13II might interfere with processes relying on an intact mitochondrial architecture. Functional studies of mitochondria with the cationic fluorochrome tetramethylrhodamine revealed that a subpopulation of the cells with p13II-positive mitochondria show a disruption in the mitochondrial inner membrane potential (Δψ), an early event observed in cells committed to apoptosis. Taken together, these results suggest novel virus-cell interactions that might be important in HTLV-I replication and/or pathogenicity.


Blood | 2011

Kinetics and intracellular compartmentalization of HTLV-1 gene expression: Nuclear retention of HBZ mRNAs

Francesca Rende; Ilaria Cavallari; Alberto Corradin; Micol Silic-Benussi; Gianna Toffolo; Yuetsu Tanaka; Steven Jacobson; Graham P. Taylor; Donna M. D'Agostino; Charles R. M. Bangham; Vincenzo Ciminale

Human T-cell leukemia virus type 1 (HTLV-1) codes for 9 alternatively spliced transcripts and 2 major regulatory proteins named Tax and Rex that function at the transcriptional and posttranscriptional levels, respectively. We investigated the temporal sequence of HTLV-1 gene expression in primary cells from infected patients using splice site-specific quantitative RT-PCR. The results indicated a two-phase kinetics with the tax/rex mRNA preceding expression of other viral transcripts. Analysis of mRNA compartmentalization in cells transfected with HTLV-1 molecular clones demonstrated the strict Rex-dependency of the two-phase kinetics and revealed strong nuclear retention of HBZ mRNAs, supporting their function as noncoding transcripts. Mathematical modeling underscored the importance of a delay between the functions of Tax and Rex, which was supported by experimental evidence of the longer half-life of Rex. These data provide evidence for a temporal pattern of HTLV-1 expression and reveal major differences in the intracellular compartmentalization of HTLV-1 transcripts.


Journal of Virology | 2002

Oncoviral bovine leukemia virus G4 and human T-cell leukemia virus type 1 p13(II) accessory proteins interact with farnesyl pyrophosphate synthetase.

Laurent Lefèbvre; Alain Vanderplasschen; Vincenzo Ciminale; Hubertine Heremans; Olivier Dangoisse; Jean Claude Jauniaux; Jean-François Toussaint; Vlado Zelnik; Arsène Burny; Richard Kettmann; Lucas Willems

ABSTRACT G4 and p13II are accessory proteins encoded by the X region of bovine leukemia virus and human T-cell leukemia virus type 1 (HTLV-1), respectively. Disruption of the G4 and p13II open reading frames interferes with viral spread in animal model systems, indicating that the corresponding proteins play a key role in viral replication. In addition, G4 is oncogenic in primary cell cultures and is absolutely required for efficient onset of leukemogenesis in sheep. To gain insight into the function of these proteins, we utilized the yeast two-hybrid system to identify protein partners of G4. Results revealed that G4 interacts with farnesyl pyrophosphate synthetase (FPPS), a protein involved in the mevalonate/squalene pathway and in synthesis of FPP, a substrate required for prenylation of Ras. The specificity of the interaction was verified by glutathione S-transferase (GST) pull-down assays and by coimmunoprecipitation experiments. Furthermore, confocal microscopy showed that the subcellular localization of G4 was profoundly affected by FPPS. The G4 protein itself was not prenylated, at least in rabbit reticulocyte lysate-based assays. The domain of G4 required for binding to FPPS was restricted to an amphipathic α-helix rich in arginine residues. Subtle mutation of this α-helix abrogated G4 oncogenic potential in vitro, providing a biological relevance for FPPS-G4 complex formation in cells. Finally, HTLV-1 p13II was also found to specifically interact with FPPS (in yeast as well as in GST pull-down assays) and to colocalize with G4 in mitochondria, suggesting a functional analogy between these oncoviral accessory proteins. Identification of FPPS as a molecular partner for p13II and G4 accessory proteins opens new prospects for treatment of retrovirus-induced leukemia.


Advances in Cancer Research | 2005

Mitochondria as functional targets of proteins coded by human tumor viruses

Donna M. D'Agostino; Paolo Bernardi; Luigi Chieco-Bianchi; Vincenzo Ciminale

Molecular analyses of tumor virus-host cell interactions have provided key insights into the genes and pathways involved in neoplastic transformation. Recent studies have revealed that the human tumor viruses Epstein-Barr virus (EBV), Kaposis sarcoma-associated herpesvirus (KSHV), human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), and human T-cell leukemia virus type 1 (HTLV-1) express proteins that are targeted to mitochondria. The list of these viral proteins includes BCL-2 homologues (BHRF1 of EBV; KSBCL-2 of KSHV), an inhibitor of apoptosis (IAP) resembling Survivin (KSHV K7), proteins that alter mitochondrial ion permeability and/or membrane potential (HBV HBx, HPV E[wedge]14, HCV p7, and HTLV-1 p13(II)), and K15 of KSHV, a protein with undefined function. Consistent with the central role of mitochondria in energy production, cell death, calcium homeostasis, and redox balance, experimental evidence indicates that these proteins have profound effects on host cell physiology. In particular, the viral BCL-2 homologues BHRF1 and KSBCL-2 inhibit apoptosis triggered by a variety of stimuli. HBx, p7, E1[wedge]4, and p13(II) exert powerful effects on mitochondria either directly due to their channel-forming activity or indirectly through interactions with endogenous channels. Further investigation of these proteins and their interactions with mitochondria will provide important insights into the mechanisms of viral replication and tumorigenesis and could aid in the discovery of new targets for anti-tumor therapy.


Journal of Virology | 2006

Human T-Lymphotropic Virus Type 1 Mitochondrion-Localizing Protein p13 II Is Required for Viral Infectivity In Vivo

Hajime Hiraragi; Seung-jae Kim; Andrew J. Phipps; Micol Silic-Benussi; Vincenzo Ciminale; Lee Ratner; Patrick L. Green; Michael D. Lairmore

ABSTRACT Human T-lymphotropic virus type 1 (HTLV-1), the etiological agent of adult T-cell leukemia, encodes unique regulatory and accessory proteins in the pX region of the provirus, including the open reading frame II product p13II. p13II localizes to mitochondria, binds farnesyl pyrophosphate synthetase, an enzyme involved in posttranslational farnesylation of Ras, and alters Ras-dependent cell signaling and control of apoptosis. The role of p13II in virus infection in vivo remains undetermined. Herein, we analyzed the functional significance of p13II in HTLV-1 infection. We compared the infectivity of a human B-cell line that harbors an infectious molecular clone of HTLV-1 with a selective mutation that prevents the translation of p13II (729.ACH.p13) to the infectivity of a wild-type HTLV-1-expressing cell line (729.ACH). 729.ACH and 729.ACH.p13 producer lines had comparable infectivities for cultured rabbit peripheral blood mononuclear cells (PBMC), and the fidelity of the start codon mutation in ACH.p13 was maintained after PBMC passage. In contrast, zero of six rabbits inoculated with 729.ACH.p13 cells failed to establish viral infection, whereas six of six rabbits inoculated with wild-type HTLV-1-expressing cells (729.ACH) were infected as measured by antibody responses, proviral load, and HTLV-1 p19 matrix antigen production from ex vivo-cultured PBMC. Our data are the first to indicate that the HTLV-1 mitochondrion-localizing protein p13II has an essential biological role during the early phase of virus infection in vivo.


Journal of Virology | 2012

The Human T-Lymphotropic Virus Type 1 Tax Protein Inhibits Nonsense-Mediated mRNA Decay by Interacting with INT6/EIF3E and UPF1

Vincent Mocquet; Julia Neusiedler; Francesca Rende; David Cluet; Jean Philippe Robin; Jean Michel Terme; Madeleine Duc Dodon; Jürgen Wittmann; Christelle Morris; Hervé Le Hir; Vincenzo Ciminale; Pierre Jalinot

ABSTRACT In this report, we analyzed whether the degradation of mRNAs by the nonsense-mediated mRNA decay (NMD) pathway was affected in human T-lymphotropic virus type 1 (HTLV-1)-infected cells. This pathway was indeed strongly inhibited in C91PL, HUT102, and MT2 cells, and such an effect was also observed by the sole expression of the Tax protein in Jurkat and HeLa cells. In line with this activity, Tax binds INT6/EIF3E (here called INT6), which is a subunit of the translation initiation factor eukaryotic initiation factor 3 (eIF3) required for efficient NMD, as well as the NMD core factor upstream frameshift protein 1 (UPF1). It was also observed that Tax expression alters the morphology of processing bodies (P-bodies), the cytoplasmic structures which concentrate RNA degradation factors. The presence of UPF1 in these subcellular compartments was increased by Tax, whereas that of INT6 was decreased. In line with these effects, the level of the phosphorylated form of UPF1 was increased in the presence of Tax. Analysis of several mutants of the viral protein showed that the interaction with INT6 is necessary for NMD inhibition. The alteration of mRNA stability was observed to affect viral transcripts, such as that coding for the HTLV-1 basic leucine zipper factor (HBZ), and also several cellular mRNAs sensitive to the NMD pathway. Our data indicate that the effect of Tax on viral and cellular gene expression is not restricted to transcriptional control but can also involve posttranscriptional regulation.


FEBS Letters | 2000

Unique features of HIV-1 Rev protein phosphorylation by protein kinase CK2 (‘casein kinase-2’)

Oriano Marin; Stefania Sarno; Marco Boschetti; Mario A. Pagano; Flavio Meggio; Vincenzo Ciminale; Donna M. D'Agostino; Lorenzo A. Pinna

The HIV‐1 Rev transactivator is phosphorylated in vitro by protein kinase CK2 at two residues, Ser‐5 and Ser‐8; these sites are also phosphorylated in vivo. Here we show that the mechanism by which CK2 phosphorylates Rev is unique in several respects, notably: (i) it is fully dependent on the regulatory, β‐subunit of CK2; (ii) it relies on the integrity of an acidic stretch of CK2β which down‐regulates the phosphorylation of other substrates; (iii) it is inhibited in a dose‐dependent manner by polyamines and other polycationic effectors that normally stimulate CK2 activity. In contrast, a peptide corresponding to the amino‐terminal 26 amino acids of Rev, including the phosphoacceptor site, is readily phosphorylated by the catalytic subunit of CK2 even in the absence of the β‐subunit. These data, in conjunction with the observation that two functionally inactive derivatives of Rev with mutations in its helix‐loop‐helix motif are refractory to phosphorylation, indicate the phosphorylation of Rev by CK2 relies on conformational features of distinct regions that are also required for the transactivators biological activity.


Journal of Virology | 2005

Human T-Lymphotropic Virus Type 1 Mitochondrion-Localizing Protein p13II Sensitizes Jurkat T Cells to Ras-Mediated Apoptosis

Hajime Hiraragi; Bindhu Michael; Amrithraj Nair; Micol Silic-Benussi; Vincenzo Ciminale; Michael D. Lairmore

ABSTRACT Human T-lymphotropic virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia. In addition to typical retroviral structural and enzymatic gene products, HTLV-1 encodes unique regulatory and accessory proteins, including a singly spliced pX open reading frame II (ORF II) product, p13II. We have demonstrated that proviral clones of HTLV-1 which are mutated in pX ORF II fail to obtain typical proviral loads and antibody responses in a rabbit animal model. p13II localizes to mitochondria and reduces cell growth and tumorigenicity in mice, but its function in human lymphocytes remains undetermined. For this study, we analyzed the functional properties of Jurkat T cells expressing p13II, using both transient and stable expression vectors. Our data indicate that p13II-expressing Jurkat T cells are sensitive to caspase-dependent, ceramide- and FasL-induced apoptosis. p13II-expressing Jurkat T cells also exhibited reduced proliferation when cultured at a high density. Furthermore, preincubation of the p13II-expressing cells with a farnesyl transferase inhibitor, which blocks the posttranslational modification of Ras, markedly reduced FasL-induced apoptosis, indicating the participation of the Ras pathway in p13IIs influence on lymphocyte survival. Our data are the first to demonstrate that p13II alters Ras-mediated apoptosis in T lymphocytes, and they reveal a potential mechanism by which HTLV-1 alters lymphocyte proliferation.

Collaboration


Dive into the Vincenzo Ciminale's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Genoveffa Franchini

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge