Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G1011-20, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24699326

ABSTRACT

Pancreatic cancer is a devastating disease with a survival rate of <5%. Moreover, pancreatic cancer aggressiveness is closely related to high levels of prosurvival mediators, which can ultimately lead to rapid disease progression. One of the mechanisms that enables tumor cells to evade cellular stress and promote unhindered proliferation is the endoplasmic reticulum (ER) stress response. Disturbances in the normal functions of the ER lead to an evolutionarily conserved cell stress response, the unfolded protein response (UPR). The UPR initially compensates for damage, but it eventually triggers cell death if ER dysfunction is severe or prolonged. Triptolide, a diterpene triepoxide, has been shown to be an effective compound against pancreatic cancer. Our results show that triptolide induces the UPR by activating the PKR-like ER kinase-eukaryotic initiation factor 2α axis and the inositol-requiring enzyme 1α-X-box-binding protein 1 axis of the UPR and leads to chronic ER stress in pancreatic cancer. Our results further show that glucose-regulated protein 78 (GRP78), one of the major regulators of ER stress, is downregulated by triptolide, leading to cell death by apoptosis in MIA PaCa-2 cells and autophagy in S2-VP10 cells.


Subject(s)
Diterpenes/pharmacology , Endoplasmic Reticulum/drug effects , Pancreatic Neoplasms/metabolism , Phenanthrenes/pharmacology , Stress, Physiological/drug effects , Unfolded Protein Response/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line, Tumor , Chronic Disease , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Epoxy Compounds/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Cancer Ther ; 12(7): 1266-75, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23635652

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest malignancies, is resistant to current chemotherapies. We previously showed that triptolide inhibits PDAC cell growth in vitro and blocks metastatic spread in vivo. Triptolide downregulates HSP70, a molecular chaperone upregulated in several tumor types. This study investigates the mechanism by which triptolide inhibits HSP70. Because microRNAs (miRNA) are becoming increasingly recognized as negative regulators of gene expression, we tested whether triptolide regulates HSP70 via miRNAs. Here, we show that triptolide as well as quercetin, but not gemcitabine, upregulated miR-142-3p in PDAC cells (MIA PaCa-2, Capan-1, and S2-013). Ectopic expression of miR-142-3p inhibited cell proliferation, measured by electric cell-substrate impedance sensing, and decreased HSP70 expression, measured by real-time PCR and immunoblotting, compared with controls. We showed that miR-142-3p directly binds to the 3'UTR of HSP70, and that this interaction is important as HSP70 overexpression rescued miR-142-3p-induced cell death. We found that miR-142-3p regulates HSP70 independently of heat shock factor 1. Furthermore, Minnelide, a water-soluble prodrug of triptolide, induced the expression of miR-142-3p in vivo. This is the first description of an miRNA-mediated mechanism of HSP70 regulation in cancer, making miR-142-3p an attractive target for PDAC therapeutic intervention.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Diterpenes/pharmacology , HSP70 Heat-Shock Proteins/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/drug therapy , Phenanthrenes/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Epoxy Compounds/pharmacology , Female , Gene Expression , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , Humans , Mice , Mice, SCID , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Random Allocation , Transfection , Xenograft Model Antitumor Assays
3.
Am J Surg ; 205(4): 387-96, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23428154

ABSTRACT

BACKGROUND: Neuroblastoma is an aggressive pediatric malignancy with significant chemotherapeutic resistance. We assessed triptolide as a potential therapy. METHODS: SH-SY5Y and IMR-32 neuroblastoma cell lines were treated with triptolide. Viability, intracellular calcium, caspase activation, protein, and mRNA levels were measured. Autophagy was evaluated with confocal microscopy. Nuclear factor-kappa B (NF-κB) activation was measured using a dual luciferase assay. RESULTS: Triptolide treatment resulted in death in both cell lines within 72 hours, with sustained increases in intracellular calcium. IMR-32 cells underwent cell death by apoptosis. Conversely, light chain 3II (LC3II) protein levels were elevated in SH-SY5Y cells, which is consistent with autophagy. Confocal microscopy confirmed increased LC3 puncta in SH-SY5Y cells compared with control cells. Heat shock pathway protein and mRNA levels decreased with treatment. NF-κB assays demonstrated inhibition of tumor necrosis factor (TNF)-α-induced activity with triptolide. CONCLUSIONS: Triptolide treatment induces cell death in neuroblastoma by different mechanisms with multiple pathways targeted. Triptolide may serve a potential chemotherapeutic role in advanced cases of neuroblastoma.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Biomarkers, Tumor/antagonists & inhibitors , Diterpenes/therapeutic use , NF-kappa B/antagonists & inhibitors , Neuroblastoma/drug therapy , Phenanthrenes/therapeutic use , Antineoplastic Agents, Alkylating/pharmacology , Biomarkers, Tumor/metabolism , Blotting, Western , Calcium/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/pharmacology , Dose-Response Relationship, Drug , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Fluorescent Antibody Technique , Heat-Shock Proteins/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Neuroblastoma/metabolism , Phenanthrenes/pharmacology , Real-Time Polymerase Chain Reaction
4.
PLoS One ; 7(8): e43020, 2012.
Article in English | MEDLINE | ID: mdl-22912777

ABSTRACT

BACKGROUND: MUC1 is a type I transmembrane glycoprotein aberrantly overexpressed in various cancer cells including pancreatic cancer. The cytosolic end of MUC1 (MUC1-c) is extensively involved in a number of signaling pathways. MUC1-c is reported to inhibit apoptosis in a number of cancer cells, but the mechanism of inhibition is unclear. METHOD: Expression of MUC1-c was studied in the pancreatic cancer cell line MIAPaCa-2 at the RNA level by using qRTPCR and at the protein level by Western blotting. MUC1-c expression was inhibited either by siRNA or by a specific peptide inhibitor, GO-201. Effect of MUC1-c inhibition on viability and proliferation and lysosomal permeabilization were studied. Association of MUC1-c with HSP70 was detected by co-immunoprecipitation of MUC1-c and HSP70. Localization of MUC1-c in cellular organelles was monitored by immunofluorescence and with immuno- blotting by MUC1-c antibody after subcellular fractionation. RESULTS: Inhibition of MUC1-c by an inhibitor (GO-201) or siRNA resulted in reduced viability and reduced proliferation of pancreatic cancer cells. Furthermore, GO-201, the peptide inhibitor of MUC1-c, was effective in reducing tumor burden in pancreatic cancer mouse model. MUC1-c was also found to be associated with HSP70 in the cytosol, although a significant amount of MUC1 was also seen to be present in the lysosomes. Inhibition of MUC1 expression or activity showed an enhanced Cathepsin B activity in the cytosol, indicating lysosomal permeabilization. Therefore this study indicates that MUC1-c interacted with HSP70 in the cytosol of pancreatic cancer cells and localized to the lysosomes in these cells. Further, our results showed that MUC1-c protects pancreatic cancer cells from cell death by stabilizing lysosomes and preventing release of Cathepsin B in the cytosol.


Subject(s)
Lysosomes/metabolism , Mucin-1/metabolism , Pancreatic Neoplasms/physiopathology , Signal Transduction/physiology , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , HSP70 Heat-Shock Proteins/metabolism , Immunoprecipitation , Mice , Peptides/pharmacology , Permeability , Protein Subunits/metabolism , Real-Time Polymerase Chain Reaction
5.
J Biosci ; 36(3): 505-16, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21799262

ABSTRACT

In the trishanku (triA-) mutant of the social amoeba Dictyostelium discoideum, aggregates are smaller than usual and the spore mass is located mid-way up the stalk, not at the apex. We have monitored aggregate territory size, spore allocation and fruiting body morphology in chimaeric groups of (quasi-wild-type) Ax2 and triA- cells. Developmental canalisation breaks down in chimaeras and leads to an increase in phenotypic variation. A minority of triA- cells causes largely Ax2 aggregation streams to break up; the effect is not due to the counting factor. Most chimaeric fruiting bodies resemble those of Ax2 or triA-. Others are double-deckers with a single stalk and two spore masses, one each at the terminus and midway along the stalk. The relative number of spores belonging to the two genotypes depends both on the mixing ratio and on the fruiting body morphology. In double-deckers formed from 1:1 chimaeras, the upper spore mass has more Ax2 spores, and the lower spore mass more triA- spores, than expected. Thus, the traits under study depend partly on the cells' own genotype and partly on the phenotypes, and so genotypes, of other cells: they are both autonomous and non-autonomous. These findings strengthen the parallels between multicellular development and behaviour in social groups. Besides that, they reinforce the point that a trait can be associated with a genotype only in a specified context.


Subject(s)
Dictyostelium/physiology , Microbial Interactions/genetics , Chimera , Culture Media, Conditioned , Dictyostelium/cytology , Fruiting Bodies, Fungal/cytology , Genes, Protozoan , Genotype
6.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G948-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21330448

ABSTRACT

Several mechanisms have evolved to ensure the survival of cells under adverse conditions. The heat shock response is one such evolutionarily conserved survival mechanism. Heat shock factor-1 (HSF1) is a transcriptional regulator of the heat shock response. By the very nature of its prosurvival function, HSF1 may contribute to the pathogenesis of cancer. The current study investigates the role of HSF1 in the pathogenesis of pancreatobiliary tumors. HSF1 was downregulated in pancreatic cancer (MIA PaCa-2 and S2-013) and cholangiocarcinoma (KMBC and KMCH) cell lines by HSF1-specific small interfering RNA (siRNA). Nonsilencing siRNA was used as control. The effect of HSF1 downregulation on viability and apoptosis parameters, i.e., annexin V, terminal deoxynucleotidyl transferase dUTP-mediated nick end labeling (TUNEL), and caspase-3, was measured. To evaluate the cancer-specific effects of HSF1, the effect of HSF1 downregulation on normal human pancreatic ductal cells was also evaluated. HSF1 is abundantly expressed in human pancreatobiliary cancer cell lines, as well as in pancreatic cancer tissue, as demonstrated by Western blot and immunohistochemistry, respectively. Inhibition of HSF1 expression by the HSF1 siRNA sequences leads to time-dependent death in pancreatic and cholangiocarcinoma cell lines. Downregulation of HSF1 expression induces annexin V and TUNEL positivity and caspase-3 activation, suggesting activation of a caspase-dependent apoptotic pathway. Although caspase-3 inhibition protects against cell death induced by HSF1 expression, it does not completely prevent it, suggesting a role for caspase-independent cell death. HSF1 plays a prosurvival role in the pathogenesis of pancreatobiliary tumors. Modulation of HSF1 activity could therefore emerge as a novel therapeutic strategy for cancer treatment.


Subject(s)
Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Cholangiocarcinoma/metabolism , DNA-Binding Proteins/metabolism , Heat-Shock Response , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Annexin A5/metabolism , Apoptosis , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Heat-Shock Response/genetics , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Signal Transduction , Time Factors , Transcription Factors/genetics
7.
Autophagy ; 6(7): 997-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818166

ABSTRACT

Pancreatic cancer, the fourth leading cause of cancer-related death in the United States, is resistant to current chemotherapies. Therefore, identification of different pathways of cell death is important to develop novel therapeutics. Our previous study has shown that triptolide, a diterpene triepoxide, inhibits the growth of pancreatic cancer cells in vitro and prevents tumor growth in vivo. However, the mechanism by which triptolide kills pancreatic cancer cells was not known, hence, this study aimed at elucidating it. Our study reveals that triptolide kills diverse types of pancreatic cancer cells by two different pathways; it induces caspase-dependent apoptotic death in some cell lines and death via a caspase-independent autophagic pathway in the other cell lines tested. Triptolide-induced autophagy requires autophagy-specific genes, atg5 or beclin 1 and its inhibition results in cell death via the apoptotic pathway, whereas inhibition of both autophagy and apoptosis rescues triptolide-mediated cell death. Our study shows for the first time that induction of autophagy by triptolide has a pro-death role in pancreatic cancer cells. Since triptolide kills diverse pancreatic cancer cells by different mechanisms, it makes an attractive chemotherapeutic agent for future use against a broad spectrum of pancreatic cancers.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Autophagy/drug effects , Cell Death/drug effects , Diterpenes/pharmacology , Pancreatic Neoplasms/pathology , Phenanthrenes/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , Diterpenes/therapeutic use , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Humans , Pancreatic Neoplasms/drug therapy , Phenanthrenes/therapeutic use
8.
Gastroenterology ; 139(2): 598-608, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20434451

ABSTRACT

BACKGROUND & AIMS: Pancreatic adenocarcinoma, among the most lethal human malignancies, is resistant to current chemotherapies. We previously showed that triptolide inhibits the growth of pancreatic cancer cells in vitro and prevents tumor growth in vivo. This study investigates the mechanism by which triptolide kills pancreatic cancer cells. METHODS: Cells were treated with triptolide and viability and caspase-3 activity were measured using colorimetric assays. Annexin V, propidium iodide, and acridine orange staining were measured by flow cytometry. Immunofluorescence was used to monitor the localization of cytochrome c and Light Chain 3 (LC3) proteins. Caspase-3, Atg5, and Beclin1 levels were down-regulated by exposing cells to their respective short interfering RNA. RESULTS: We show that triptolide induces apoptosis in MiaPaCa-2, Capan-1, and BxPC-3 cells and induces autophagy in S2-013, S2-VP10, and Hs766T cells. Triptolide-induced autophagy has a pro-death effect, requires autophagy-specific genes, atg5 or beclin1, and is associated with the inactivation of the Protein kinase B (Akt)/mammalian target of Rapamycin/p70S6K pathway and the up-regulation of the Extracellular Signal-Related Kinase (ERK)1/2 pathway. Inhibition of autophagy in S2-013 and S2-VP10 cells results in cell death via the apoptotic pathway whereas inhibition of both autophagy and apoptosis rescues cell death. CONCLUSIONS: This study shows that triptolide kills pancreatic cancer cells by 2 different pathways. It induces caspase-dependent apoptotic death in MiaPaCa-2, Capan-1, and BxPC-3, and induces caspase-independent autophagic death in metastatic cell lines S2-013, S2-VP10, and Hs766T, thereby making it an attractive chemotherapeutic agent against a broad spectrum of pancreatic cancers.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Diterpenes/pharmacology , Pancreatic Neoplasms/pathology , Phenanthrenes/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epoxy Compounds/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Time Factors
9.
J Gastrointest Surg ; 14(2): 252-60, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20013316

ABSTRACT

INTRODUCTION: An emerging therapy in oncology is the induction of apoptotic cell death through anti-death receptor therapy. However, pancreatic cancer is resistant to apoptosis including anti-death receptor therapy. We have previously described how triptolide decreases resistance to apoptosis in pancreatic cancer cells in vitro and in vivo. We hypothesized that triptolide decreases tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resistance in pancreatic cancer cells. The aim of this study was to evaluate the effects that combined therapy with TRAIL and triptolide have on different parameters of apoptosis. METHODS: Four different pancreatic cancer cell lines were exposed to triptolide, TRAIL, or a combination of both drugs. We assessed the effects that combined therapy with TRAIL and triptolide has on cell viability, apoptosis, caspase-3 and caspase-9 activities, and poly(ADP)-ribose polymerase cleavage. RESULTS: Pancreatic cancer cells were resistant to TRAIL therapy; however, combined therapy with triptolide and TRAIL significantly decreased the cell viability in all the cell lines and increased apoptotic cell death as a result of caspase-3 and caspase-9 activation. CONCLUSIONS: Pancreatic cancer is highly resistant to anti-death receptor therapy, but combined therapy with TRAIL and triptolide is an effective therapy that induces apoptotic cell death in pancreatic cancer cells.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Phenanthrenes/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adenocarcinoma , Cell Line, Tumor , Cell Survival/drug effects , Epoxy Compounds/pharmacology , Humans , Pancreatic Neoplasms
10.
Evol Dev ; 11(6): 697-709, 2009.
Article in English | MEDLINE | ID: mdl-19878291

ABSTRACT

Multicellular development in the social amoeba Dictyostelium discoideum is triggered by starvation. It involves a series of morphogenetic movements, among them being the rising of the spore mass to the tip of the stalk. The process requires precise coordination between two distinct cell types-presumptive (pre-) spore cells and presumptive (pre-) stalk cells. Trishanku (triA) is a gene expressed in prespore cells that is required for normal morphogenesis. The triA(-) mutant shows pleiotropic effects that include an inability of the spore mass to go all the way to the top. We have examined the cellular behavior required for the normal ascent of the spore mass. Grafting and mixing experiments carried out with tissue fragments and cells show that the upper cup, a tissue that derives from prestalk cells and anterior-like cells (ALCs), does not develop properly in a triA(-) background. A mutant upper cup is unable to lift the spore mass to the top of the fruiting body, likely due to defective intercellular adhesion. If wild-type upper cup function is provided by prestalk and ALCs, trishanku spores ascend all the way. Conversely, Ax2 spores fail to do so in chimeras in which the upper cup is largely made up of mutant cells. Besides proving that under these conditions the wild-type phenotype of the upper cup is necessary and sufficient for terminal morphogenesis in D. discoideum, this study provides novel insights into developmental and evolutionary aspects of morphogenesis in general. Genes that are active exclusively in one cell type can elicit behavior in a second cell type that enhances the reproductive fitness of the first cell type, thereby showing that morphogenesis is a cooperative process.


Subject(s)
Cell Differentiation/genetics , Dictyostelium/physiology , Gene Expression Regulation, Developmental , Genes, Protozoan/physiology , Animals , Cell Aggregation/genetics , Genes, Protozoan/genetics , Green Fluorescent Proteins/metabolism , Models, Biological , Models, Genetic , Morphogenesis/genetics , Phenotype
11.
Gastroenterology ; 136(5): 1772-82, 2009 May.
Article in English | MEDLINE | ID: mdl-19208367

ABSTRACT

BACKGROUND & AIMS: Heat shock proteins (HSPs) are highly conserved and serve a multitude of functions that mediate cell survival. HSP70, the only inducible form of the 70-kilodalton subfamily of HSPs, is overexpressed in pancreatic cancer cells and has been shown to inhibit caspase-dependent apoptosis. We aimed to elucidate the mechanism by which HSP70 inhibits apoptosis in cancer cells. METHODS: HSP70 expression was down-regulated in cultured pancreatic cancer cells by exposure to quercetin, triptolide, or short interfering RNAs. Intracellular Ca2+, cytosolic cathepsin B activity, caspase-3 activity, cell viability, and lysosome integrity were measured using colorimetric assays. Immunofluorescence assays were used to localize cathepsin B and Lamp2. BAPTA-AM was used to chelate intracellular Ca2+. RESULTS: Inhibition of HSP70 increased intracellular Ca2+ levels in pancreatic and colon cancer cell lines and led to loss of lysosome integrity in pancreatic cancer cells. The release of intracellular Ca2+ and lysosomal enzymes activated caspase-dependent apoptosis independently and simultaneously. CONCLUSIONS: HSP70 inhibits apoptosis in cancer cells by 2 mechanisms: attenuation of cytosolic calcium and stabilization of lysosomes. HSP70-mediated cell survival might occur in other types of cancer cells.


Subject(s)
Apoptosis/drug effects , HSP70 Heat-Shock Proteins/pharmacology , Pancreatic Neoplasms/pathology , Calcium/metabolism , Caspase 3/metabolism , Cathepsin B/metabolism , Cell Line, Tumor , Cytosol/metabolism , Diterpenes/pharmacology , Epoxy Compounds/pharmacology , Humans , Lysosomal-Associated Membrane Protein 2/pharmacology , Lysosomes/drug effects , Pancreatic Neoplasms/metabolism , Phenanthrenes/pharmacology , Quercetin/pharmacology , RNA, Small Interfering/pharmacology
12.
Differentiation ; 74(9-10): 596-607, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17177856

ABSTRACT

We have identified a novel gene, trishanku (triA), by random insertional mutagenesis of Dictyostelium discoideum. TriA is a Broad complex Tramtrack bric-a-brac domain-containing protein that is expressed strongly during the late G2 phase of cell cycle and in presumptive spore (prespore (psp)) cells. Disrupting triA destabilizes cell fate and reduces aggregate size; the fruiting body has a thick stalk, a lowered spore: stalk ratio, a sub-terminal spore mass and small, rounded spores. These changes revert when the wild-type triA gene is re-expressed under a constitutive or a psp-specific promoter. By using short- and long-lived reporter proteins, we show that in triA(-) slugs the prestalk (pst)/psp proportion is normal, but that there is inappropriate transdifferentiation between the two cell types. During culmination, regardless of their current fate, all cells with a history of pst gene expression contribute to the stalk, which could account for the altered cell-type proportion in the mutant.


Subject(s)
Dictyostelium/growth & development , Morphogenesis , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Cell Cycle , Cell Differentiation , Cloning, Molecular , Dictyostelium/genetics , Dictyostelium/physiology , Gene Expression Regulation, Developmental , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Phenotype , Protozoan Proteins/metabolism , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...