Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 137(6): 1318-29, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25716227

ABSTRACT

The ability to escape apoptosis is a hallmark of cancer-initiating cells and a key factor of resistance to oncolytic therapy. Here, we identify FAM96A as a ubiquitous, evolutionarily conserved apoptosome-activating protein and investigate its potential pro-apoptotic tumor suppressor function in gastrointestinal stromal tumors (GISTs). Interaction between FAM96A and apoptotic peptidase activating factor 1 (APAF1) was identified in yeast two-hybrid screen and further studied by deletion mutants, glutathione-S-transferase pull-down, co-immunoprecipitation and immunofluorescence. Effects of FAM96A overexpression and knock-down on apoptosis sensitivity were examined in cancer cells and zebrafish embryos. Expression of FAM96A in GISTs and histogenetically related cells including interstitial cells of Cajal (ICCs), "fibroblast-like cells" (FLCs) and ICC stem cells (ICC-SCs) was investigated by Northern blotting, reverse transcription-polymerase chain reaction, immunohistochemistry and Western immunoblotting. Tumorigenicity of GIST cells and transformed murine ICC-SCs stably transduced to re-express FAM96A was studied by xeno- and allografting into immunocompromised mice. FAM96A was found to bind APAF1 and to enhance the induction of mitochondrial apoptosis. FAM96A protein or mRNA was dramatically reduced or lost in 106 of 108 GIST samples representing three independent patient cohorts. Whereas ICCs, ICC-SCs and FLCs, the presumed normal counterparts of GIST, were found to robustly express FAM96A protein and mRNA, FAM96A expression was much reduced in tumorigenic ICC-SCs. Re-expression of FAM96A in GIST cells and transformed ICC-SCs increased apoptosis sensitivity and diminished tumorigenicity. Our data suggest FAM96A is a novel pro-apoptotic tumor suppressor that is lost during GIST tumorigenesis.


Subject(s)
Apoptosis/genetics , Carrier Proteins/genetics , Gastrointestinal Stromal Tumors/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptotic Protease-Activating Factor 1/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression/genetics , HEK293 Cells , Humans , Interstitial Cells of Cajal/metabolism , Metalloproteins , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Mitochondria/genetics , Zebrafish/genetics
2.
Nucleic Acid Ther ; 21(6): 423-36, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22196370

ABSTRACT

The toll-like receptors (TLRs) 7, 8, and 9 stimulate innate immune responses upon recognizing pathogen nucleic acids. Certain GU- or AU-rich RNA sequences were described to differentiate between human TLR7- and TLR8-mediated immune effects. Those single-stranded RNA molecules require endosomal delivery for stabilization against ribonucleases. We have discovered RNA sequences that preferentially activate TLR7, form higher ordered structures, and do not require specific cellular delivery. In addition, a dual activation of TLR8 and TLR9 without affecting TLR7 can be achieved by chimeric molecules consisting of GU-rich RNA and Cytosin (C) phosphordiester or phosphorthioat (p) guanine (CpG) motif DNA sequences. Such chimeras stimulate TLR9-mediated type I interferon (IFN) and TLR8-depending proinflammatory cytokine and chemokine production upon primary human cell activation. However, an RNA-dependent TLR7 IFN-α cytokine release is suppressed by the phosphorothioate DNA sequence contained in the chimeric molecule. To convert the immune response of a single-stranded RNA from TLR7/8 to TLR9, a simple chemical modification at the 5' end proves to be sufficient. Such 8-oxo-2'-deoxy-guanosine or 8-bromo-2'-deoxy-guanosine modifications of the first guanosine in GU-rich single-stranded RNAs convert the immune response to include TLR9 activation and demonstrate strong additive effects for type I IFN immune responses in human primary cells.


Subject(s)
Oligoribonucleotides/administration & dosage , Oligoribonucleotides/chemistry , Toll-Like Receptor 7/drug effects , Toll-Like Receptor 8/drug effects , Toll-Like Receptor 9/drug effects , Animals , Cells, Cultured , Chemokines/drug effects , Cytokines/drug effects , Female , HEK293 Cells/drug effects , HEK293 Cells/immunology , Humans , Immunity, Innate/drug effects , Interferon Type I/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Mice , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemistry , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Toll-Like Receptor 9/immunology
3.
Oligonucleotides ; 17(4): 405-17, 2007.
Article in English | MEDLINE | ID: mdl-18072859

ABSTRACT

Viruses of the order Mononegavirales encompass life-threatening pathogens with single-stranded segmented or nonsegmented negative-strand RNA genomes. The RNA genomes are characterized by highly conserved sequences at the extreme untranslated 3' and 5' termini that are most important for virus infection and viral RNA synthetic processes. The 3' terminal genome regions of negative-strand viruses such as vesicular stomatitis virus, Sendai virus, or influenza virus contain a high number of conserved U and G nucleotides, and synthetic oligoribonucleotides encoding such sequences stimulate sequence-dependent cytokine responses via TLR7 and TLR8. Immune cells responding to such sequences include NK cells, NK/T cells, plasmacytoid, and myeloid dendritic cells, as well as monocytes and B cells. Strong Th1 and pro-inflammatory cytokine responses are also induced upon in vivo application of oligoribonucleotides. It appears possible that the presence of highly conserved untranslated terminal regions in the viral genome fulfilling fundamental functions for the viral replication may enable the host to induce directed innate immune defense mechanisms, by allowing pathogen detection through essential RNA regions that the virus cannot readily mutate.


Subject(s)
Immunity, Innate , Mononegavirales/immunology , Oligodeoxyribonucleotides/immunology , Oligoribonucleotides/immunology , RNA, Viral/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Animals , Cell Line , Conserved Sequence , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Genome, Viral , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/immunology , Mononegavirales/metabolism , Oligodeoxyribonucleotides/pharmacology , Oligoribonucleotides/pharmacology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology
4.
FASEB J ; 17(10): 1295-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12759333

ABSTRACT

Apoptosis is a fundamental biological process used to eliminate unwanted cells in a multicellular organism. An increasing number of regulatory proteins have been identified that either promote or inhibit apoptosis. For tumors to arise, apoptosis must be blocked in the transformed cells, for example by mutational overexpression of anti-apoptotic proteins, which represent attractive target proteins for molecular therapy strategies. In a functional yeast survival screen designed to select new anti-apoptotic mammalian genes, we have identified the chromosomal high-mobility group box-1 protein (HMGB1) as an inhibitor of yeast cell death induced by the pro-apoptotic Bcl-2 family member Bak. The C-terminal 33 amino acids of HMGB1 are dispensable for this inhibitory function. HMGB1 is also able to protect mammalian cells against different death stimuli including ultraviolet radiation, CD95-, TRAIL-, Casp-8-, and Bax-induced apoptosis. We found high HMGB1 protein levels in human primary breast carcinoma. Hmgb1 RNA levels are changing during different stages of mouse mammary gland development and are particularly low during lactation and involution. These data suggest that HMGB1 may participate in the regulation of mammary gland apoptosis and that its high expression level promotes tumor growth because of its anti-apoptotic properties.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Carcinoma/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/physiology , Animals , Cell Line , Cytoprotection , Female , Gene Expression Regulation, Developmental , HMGB1 Protein/genetics , Humans , Mammary Glands, Animal/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Models, Biological , RNA, Messenger/metabolism , Rats , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics , Transformation, Genetic , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...