Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 88(10): 5263-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24574398

ABSTRACT

UNLABELLED: Novel therapies employing oncolytic viruses have emerged as promising anticancer modalities. The cure of particularly aggressive malignancies requires induction of immunogenic cell death (ICD), coupling oncolysis with immune responses via calreticulin, ATP, and high-mobility group box protein B1 (HMGB1) release from dying tumor cells. The present study shows that in human pancreatic cancer cells (pancreatic ductal adenocarcinoma [PDAC] cells n=4), oncolytic parvovirus H-1 (H-1PV) activated multiple interconnected death pathways but failed to induce calreticulin exposure or ATP release. In contrast, H-1PV elevated extracellular HMGB1 levels by 4.0±0.5 times (58%±9% of total content; up to 100 ng/ml) in all infected cultures, whether nondying, necrotic, or apoptotic. An alternative secretory route allowed H-1PV to overcome the failure of gemcitabine to trigger HMGB1 release, without impeding cytotoxicity or other ICD activities of the standard PDAC medication. Such broad resistance of H-1PV-induced HMGB1 release to apoptotic blockage coincided with but was uncoupled from an autocrine interleukin-1ß (IL-1ß) loop. That and the pattern of viral determinants maintained in gemcitabine-treated cells suggested the activation of an inflammasome/caspase 1 (CASP1) platform alongside DNA detachment and/or nuclear exclusion of HMGB1 during early stages of the viral life cycle. We concluded that H-1PV infection of PDAC cells is signaled through secretion of the alarmin HMGB1 and, besides its own oncolytic effect, might convert drug-induced apoptosis into an ICD process. A transient arrest of cells in the cyclin A1-rich S phase would suffice to support compatibility of proliferation-dependent H-1PV with cytotoxic regimens. These properties warrant incorporation of the oncolytic virus H-1PV, which is not pathogenic in humans, into multimodal anticancer treatments. IMPORTANCE: The current therapeutic concepts targeting aggressive malignancies require an induction of immunogenic cell death characterized by exposure of calreticulin (CRT) as well as release of ATP and HMGB1 from dying cells. In pancreatic tumor cells (PDAC cells) infected with the oncolytic parvovirus H-1PV, only HMGB1 was released by all infected cells, whether nondying, necrotic, or succumbing to one of the programmed death pathways, including contraproductive apoptosis. Our data suggest that active secretion of HMGB1 from PDAC cells is a sentinel reaction emerging during early stages of the viral life cycle, irrespective of cell death, that is compatible with and complements cytotoxic regimens. Consistent induction of HMGB1 secretion raised the possibility that this reaction might be a general "alarming" phenomenon characteristic of H-1PV's interaction with the host cell; release of IL-1ß points to the possible involvement of a danger-sensing inflammasome platform. Both provide a basis for further virus-oriented studies.


Subject(s)
Antineoplastic Agents/metabolism , Cell Death , Deoxycytidine/analogs & derivatives , Epithelial Cells/physiology , Oncolytic Viruses/growth & development , Parvovirus/growth & development , Cell Line, Tumor , Deoxycytidine/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , HMGB1 Protein/metabolism , Humans , Signal Transduction , Gemcitabine
2.
Cancer Biol Ther ; 10(12): 1280-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21124075

ABSTRACT

Treatment of cancers by means of viruses, that specifically replicate in (oncotropism) and kill (oncolysis) neoplastic cells, is increasingly gaining acceptance in the clinic. Among these agents, parvoviruses have been shown to possess not only direct oncolytic but also immunomodulating properties, serving as an adjuvant to prime the immune system to react against infected tumors. Here, we aimed to establish whether immunomodulating mechanisms participate in the recently reported therapeutic potential of parvoviruses against pancreatic carcinoma. Using adoptive transfer experiments we discovered that the transfer of splenocytes of donor rats harboring H-1PV-treated orthotopic PDAC tumors could significantly prolong the survival of naïve tumor-bearing recipients, compared to those receiving cells from mock-treated donors. Closer investigation of immunological parameters in infected donor rats revealed that virus-induced interferon gamma production and cellular immune response played an important role in this effect. These data have also preclinical relevance since abortive H-1PV infection of human peripheral blood mononuclear cells or cocultivation of these cells with H-1PV-preinfected pancreatic cancer cells, resulted in enhancement of innate and adaptive immune reactivity. Taken together our data reveal that oncolytic H-1PV modulates the immune system into an anticancer state, and further support the concept of using parvoviruses in the fight against pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , H-1 parvovirus/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Oncolytic Virotherapy/methods , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/metabolism , Flow Cytometry , H-1 parvovirus/physiology , Humans , Immunity, Cellular/immunology , Immunomodulation , Interferon-gamma/immunology , Oncolytic Viruses , Polymerase Chain Reaction , Rats , Spleen/immunology , Spleen/virology , Th1-Th2 Balance
3.
J Virol ; 84(1): 516-31, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19864388

ABSTRACT

Parvovirus minute virus of mice (MVMp) is endowed with oncotropic properties so far ascribed only to the dependency of the virus life cycle on cellular factors expressed during S phase and/or modulated by malignant transformation. For other viruses oncotropism relies on their inability to circumvent type I interferon (IFN)-induced innate antiviral mechanisms, the first line of defense triggered by normal cells against viral infections. These agents propagate, therefore, preferentially in transformed/tumor cells, which often lack functional antiviral mechanisms. The present study aimed at investigating whether antiviral processes also contribute to MVMp oncotropism. Our results demonstrate that in contrast to MVMp-permissive transformed mouse A9 fibroblasts, freshly isolated normal counterparts (mouse embryonic fibroblasts [MEFs]) mount, through production and release of type I IFNs upon their infection, an antiviral response against MVMp lytic multiplication. Pretreatment of MEFs with a type I IFN-beta-neutralizing antibody, prior to MVMp infection, inhibits the virus-triggered antiviral response and improves the fulfillment of the MVMp life cycle. Our results also show that part of the A9 permissiveness to MVMp relies on the inability to produce type I IFNs upon parvovirus infection, a feature related either to an A9 intrinsic deficiency of this process or to an MVMp-triggered inhibitory mechanism, since stimulation of these cells by exogenous IFN-beta strongly inhibits the parvovirus life cycle. Taken together, our results demonstrate for the first time that parvovirus infection triggers an innate antiviral response in normal cells and suggest that the MVMp oncotropism depends at least in part on the failure of infected transformed cells to mount such a response.


Subject(s)
Immunity, Innate , Minute Virus of Mice/immunology , Animals , Cell Line, Transformed , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/virology , Humans , Interferon Type I/biosynthesis , Interferon-beta/pharmacology , Mice , Virus Replication/drug effects
4.
Biochim Biophys Acta ; 1773(2): 109-19, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17034881

ABSTRACT

Cysteine string proteins (CSPs) belong to the DnaJ-like chaperone family and play an important role in regulated exocytosis in neurons and endocrine cells. The palmitoylation of several residues in a cysteine string domain may anchor CSPs to the exocytotic vesicle surface and in pancreatic beta-cells, Cspalpha is localized on insulin containing large dense core vesicles (LDCVs). An isoform closely related to Cspalpha, Cspbeta, has been obtained from testis cell cDNA libraries. To gain insights on this isoform and more generally on the properties of CSPs, we compared Cspalpha and Cspbeta. In pull-down experiments, Cspbeta was able to interact to the same extent with two of the known Cspalpha chaperone partners, Hsc70 and SGT. Upon transient overexpression in clonal beta-cells, Cspbeta but not Cspalpha was mainly produced as a non-palmitoylated protein and mutational analysis indicated that domains distinct from the cysteine string are responsible for this difference. As Cspbeta remained tightly bound to membranes, intrinsic properties of CSPs are sufficient for interactions with membranes. Indeed, recombinant Cspalpha and Cspbeta were capable to interact with membranes even in their non-palmitoylated forms. Furthermore, overexpressed Cspbeta was not associated with LDCVs, but was localized at the trans-Golgi network. Our results suggest a possible correlation between the specific membrane targeting and the palmitoylation level of CSPs.


Subject(s)
HSP40 Heat-Shock Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Membrane Proteins/metabolism , Palmitic Acid/metabolism , trans-Golgi Network/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Membrane/metabolism , Clone Cells/metabolism , DNA, Complementary/genetics , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , HSC70 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
Exp Cell Res ; 312(13): 2500-14, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16777091

ABSTRACT

The human small glutamine-rich TPR-containing protein (hSGT) is essential for cell division since RNA-interference-mediated strong reduction of hSGT protein levels causes mitotic arrest (M. Winnefeld, J. Rommelaere, and C. Cziepluch, The human small glutamine-rich TPR-containing protein is required for progress through cell division, Exp. Cell Res. 293 (2004), 43-57). Analysis of HeLa cells expressing a histone 2A-YFP fusion protein revealed the continuous presence of few mislocalized chromosomes close to the spindle poles as possible cause for hSGT depletion-dependent prometaphase arrest. Cells unable to rescue these mislocalized chromosomes into the metaphase plate died at this stage through apoptosis. In order to address hSGT function at the molecular level, mass spectrometry analysis of proteins which co-immunoprecipitated with Flag-tagged hSGT was performed. Thereby, Hsp70 and Bag-6/Bat-3/Scythe were identified as novel hSGT interaction partners while interaction with Hsc70 was confirmed. Results obtained with truncated versions of the hSGT protein revealed that Bag-6/Bat-3/Scythe and Hsp70 or Hsc70 were independently able to form complexes with hSGT. Interaction of hSGT with Hsc70, Hsp70 or Bag-6/Bat-3/Scythe was demonstrated in prometaphase, thereby suggesting a possible role for complexes containing hSGT and distinct (co)-chaperones during mitosis. Finally, cells from populations with reduced levels of Bag-6/Bat-3/Scythe also displayed persistence of mislocalized chromosomes and mitotic arrest, which strongly indicated that hSGT-Bag-6/Bat-3/Scythe complexes could be directly or indirectly required for complete chromosome congression.


Subject(s)
Carrier Proteins/metabolism , Chromosome Pairing , Chromosomes, Human/metabolism , Mitosis , Proteins/metabolism , Apoptosis , Binding Sites/genetics , Carrier Proteins/chemistry , Epithelial Cells/cytology , HSC70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Molecular Chaperones , Protein Binding , Recombinant Fusion Proteins/metabolism , Spindle Apparatus/metabolism , Tumor Cells, Cultured
6.
Exp Cell Res ; 293(1): 43-57, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14729056

ABSTRACT

Eukaryotic organisms from yeast to human harbor genes encoding the small glutamine-rich tetratricopeptide repeat-containing (SGT) protein. Work presented here demonstrated the presence of human SGT (hSGT) protein in a panel of human cell lines and throughout the cell cycle. To identify cellular processes in which hSGT is involved, knock down populations were analyzed which were generated through transfection of hsgt-specific small interfering RNA. Most strikingly, depletion of hSGT led to reduced proliferation of the affected cell populations while the mitotic index was increased. Time-lapse video microscopy revealed that cells from hSGT-depleted populations were unable to complete cell division due to mitotic arrest which was frequently followed by cell death. Further evidence for a role in cell division was given by the accumulation of hSGT in the midzone and the midbody, and by a mitosis-specific migration pattern of hSGT as detected by Western blotting after SDS-PAGE or two-dimensional gel electrophoresis. In conclusion, results obtained in this study demonstrate that hSGT protein is a constitutive component of all human cell lines tested and that this protein is essential for successful completion of cell division.


Subject(s)
Cell Division , Glutamine/chemistry , Glutamine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoptosis , Cell Line , HeLa Cells , Humans , Mitosis , RNA, Small Interfering/metabolism , Time Factors , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...