Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(19): 2704-2714, 2017 05 11.
Article in English | MEDLINE | ID: mdl-27991927

ABSTRACT

Caspase-2, one of the most evolutionarily conserved of the caspase family, has been implicated in maintenance of chromosomal stability and tumour suppression. Caspase-2 deficient (Casp2-/-) mice develop normally but show premature ageing-related traits and when challenged by certain stressors, succumb to enhanced tumour development and aneuploidy. To test how caspase-2 protects against chromosomal instability, we utilized an ex vivo system for aneuploidy where primary splenocytes from Casp2-/- mice were exposed to anti-mitotic drugs and followed up by live cell imaging. Our data show that caspase-2 is required for deleting mitotically aberrant cells. Acute silencing of caspase-2 in cultured human cells recapitulated these results. We further generated Casp2C320S mutant mice to demonstrate that caspase-2 catalytic activity is essential for its function in limiting aneuploidy. Our results provide direct evidence that the apoptotic activity of caspase-2 is necessary for deleting cells with mitotic aberrations to limit aneuploidy.


Subject(s)
Aneuploidy , Apoptosis/genetics , Caspase 2/genetics , Chromosomal Instability/genetics , Animals , Caspase 2/metabolism , Humans , Mice , Mice, Knockout , Oxidative Stress/genetics
2.
Cell Death Discov ; 2: 16009, 2016.
Article in English | MEDLINE | ID: mdl-27551503

ABSTRACT

Gender-specific differences are commonly found in metabolic pathways and in response to nutritional manipulation. Previously, we identified a role for caspase-2 in age-related glucose homeostasis and lipid metabolism using male caspase-2-deficient (Casp2 (-/-) ) mice. Here we show that the resistance to age-induced glucose tolerance does not occur in female Casp2 (-/-) mice and it appears to be independent of insulin sensitivity in males. Using fasting (18 h) as a means to further investigate the role of caspase-2 in energy and lipid metabolism, we identified sex-specific differences in the fasting response and lipid mobilization. In aged (18-22 months) male Casp2 (-/-) mice, a significant decrease in fasting liver mass, but not total body weight, was observed while in females, total body weight, but not liver mass, was reduced when compared with wild-type (WT) animals. Fasting-induced lipolysis of adipose tissue was enhanced in male Casp2 (-/-) mice as indicated by a significant reduction in white adipocyte cell size, and increased serum-free fatty acids. In females, white adipocyte cell size was significantly smaller in both fed and fasted Casp2 (-/-) mice. No difference in fasting-induced hepatosteatosis was observed in the absence of caspase-2. Further analysis of white adipose tissue (WAT) indicated that female Casp2 (-/-) mice may have enhanced fatty acid recycling and metabolism with expression of genes involved in glyceroneogenesis and fatty acid oxidation increased. Loss of Casp2 also increased fasting-induced autophagy in both male and female liver and in female skeletal muscle. Our observations suggest that caspase-2 can regulate glucose homeostasis and lipid metabolism in a tissue and sex-specific manner.

3.
Cell Death Differ ; 23(10): 1727-36, 2016 10.
Article in English | MEDLINE | ID: mdl-27518436

ABSTRACT

Aberrant cell death/survival has a critical role in the development of hepatocellular carcinoma (HCC). Caspase-2, a cell death protease, limits oxidative stress and chromosomal instability. To study its role in reactive oxygen species (ROS) and DNA damage-induced liver cancer, we assessed diethylnitrosamine (DEN)-mediated tumour development in caspase-2-deficient (Casp2(-/-)) mice. Following DEN injection in young animals, tumour development was monitored for 10 months. We found that DEN-treated Casp2(-/-) mice have dramatically elevated tumour burden and accelerated tumour progression with increased incidence of HCC, accompanied by higher oxidative damage and inflammation. Furthermore, following acute DEN injection, liver injury, DNA damage, inflammatory cytokine release and hepatocyte proliferation were enhanced in mice lacking caspase-2. Our study demonstrates for the first time that caspase-2 limits the progression of tumourigenesis induced by an ROS producing and DNA damaging reagent. Our findings suggest that after initial DEN-induced DNA damage, caspase-2 may remove aberrant cells to limit liver damage and disease progression. We propose that Casp2(-/-) mice, which are more susceptible to genomic instability, are limited in their ability to respond to DNA damage and thus carry more damaged cells resulting in accelerated tumourigenesis.


Subject(s)
Caspase 2/deficiency , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Caspase 2/metabolism , Cell Death , Cell Proliferation , DNA Damage , Diethylnitrosamine , Enzyme Activation , Inflammation/complications , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/pathology , Liver Neoplasms/enzymology , Male , Mice, Inbred C57BL , Neoplasm Staging , Oxidative Stress , Stress, Physiological
5.
Cell Death Differ ; 22(11): 1803-11, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25857265

ABSTRACT

The receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD/CRADD) functions as a dual adaptor and is a constituent of different multi-protein complexes implicated in the regulation of inflammation and cell death. Within the PIDDosome complex, RAIDD connects the cell death-related protease, Caspase-2, with the p53-induced protein with a death domain 1 (PIDD1). As such, RAIDD has been implicated in DNA-damage-induced apoptosis as well as in tumorigenesis. As loss of Caspase-2 leads to an acceleration of tumor onset in the Eµ-Myc mouse lymphoma model, whereas loss of Pidd1 actually delays onset of this disease, we set out to interrogate the role of Raidd in cancer in more detail. Our data obtained analyzing Eµ-Myc/Raidd(-/-) mice indicate that Raidd is unable to protect from c-Myc-driven lymphomagenesis. Similarly, we failed to observe a modulatory effect of Raidd deficiency on DNA-damage-driven cancer. The role of Caspase-2 as a tumor suppressor and that of Pidd1 as a tumor promoter can therefore be uncoupled from their ability to interact with the Raidd scaffold, pointing toward the existence of alternative signaling modules engaging these two proteins in this context.


Subject(s)
CRADD Signaling Adaptor Protein/metabolism , Caspase 2/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , CRADD Signaling Adaptor Protein/genetics , Caspase 2/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , Death Domain Receptor Signaling Adaptor Proteins/genetics , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Mice , Proto-Oncogene Proteins c-myc/genetics
6.
Cell Death Dis ; 6: e1615, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25611376

ABSTRACT

Ageing is a complex biological process for which underlying biochemical changes are still largely unknown. We performed comparative profiling of the cellular proteome and metabolome to understand the molecular basis of ageing in Caspase-2-deficient (Casp2(-/-)) mice that are a model of premature ageing in the absence of overt disease. Age-related changes were determined in the liver and serum of young (6-9 week) and aged (18-24 month) wild-type and Casp2(-/-) mice. We identified perturbed metabolic pathways, decreased levels of ribosomal and respiratory complex proteins and altered mitochondrial function that contribute to premature ageing in the Casp2(-/-) mice. We show that the metabolic profile changes in the young Casp2(-/-) mice resemble those found in aged wild-type mice. Intriguingly, aged Casp2(-/-) mice were found to have reduced blood glucose and improved glucose tolerance. These results demonstrate an important role for caspase-2 in regulating proteome and metabolome remodelling during ageing.


Subject(s)
Aging/metabolism , Caspase 2/deficiency , Metabolome , Proteome/metabolism , Aging/blood , Amino Acids/metabolism , Animals , Caspase 2/metabolism , Glucose/metabolism , Glucose Intolerance , Homeostasis , Lipid Metabolism , Liver/metabolism , Male , Metabolomics , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NADP/metabolism , Oxidative Phosphorylation , Pentose Phosphate Pathway , Proteomics , Reproducibility of Results , Signal Transduction
7.
Cell Death Differ ; 22(4): 526-39, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25526085

ABSTRACT

Caspases are proteases with a well-defined role in apoptosis. However, increasing evidence indicates multiple functions of caspases outside apoptosis. Caspase-1 and caspase-11 have roles in inflammation and mediating inflammatory cell death by pyroptosis. Similarly, caspase-8 has dual role in cell death, mediating both receptor-mediated apoptosis and in its absence, necroptosis. Caspase-8 also functions in maintenance and homeostasis of the adult T-cell population. Caspase-3 has important roles in tissue differentiation, regeneration and neural development in ways that are distinct and do not involve any apoptotic activity. Several other caspases have demonstrated anti-tumor roles. Notable among them are caspase-2, -8 and -14. However, increased caspase-2 and -8 expression in certain types of tumor has also been linked to promoting tumorigenesis. Increased levels of caspase-3 in tumor cells causes apoptosis and secretion of paracrine factors that promotes compensatory proliferation in surrounding normal tissues, tumor cell repopulation and presents a barrier for effective therapeutic strategies. Besides this caspase-2 has emerged as a unique caspase with potential roles in maintaining genomic stability, metabolism, autophagy and aging. The present review focuses on some of these less studied and emerging functions of mammalian caspases.


Subject(s)
Caspases/metabolism , Animals , Apoptosis , Carcinogenesis , Caspases/chemistry , Cell Differentiation , Genomic Instability , Humans , Inflammation Mediators/metabolism , Regeneration
8.
Oncogene ; 34(38): 4995-5002, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-25531319

ABSTRACT

Caspase-2 belongs to the caspase family of cysteine proteases with established roles in apoptosis. Recently, caspase-2 has been implicated in nonapoptotic functions including maintenance of genomic stability and tumor suppression. Our previous studies demonstrated that caspase-2 also regulates cellular redox status and delays the onset of several ageing-related traits. In the current study, we tested stress tolerance ability in caspase-2-deficient (Casp2(-/-)) mice by challenging both young and old mice with a low dose of the potent reactive oxygen species (ROS) generator, PQ that primarily affects lungs. In both groups of mice, PQ induced pulmonary damage. However, the lesions in caspase-2 knockout mice were consistently and reproducibly more severe than those in wild-type (WT) mice. Furthermore, serum interleukin (IL)-1ß and IL-6 levels were higher in PQ-exposed aged Casp2(-/-) mice indicating increased inflammation. Interestingly, livers from Casp2(-/-) mice displayed karyomegaly, a feature commonly associated with ageing and aneuploidy. Given that Casp2(-/-) mice show impaired antioxidant defense, we tested oxidative damage in these mice. Protein oxidation significantly increased in PQ-injected old Casp2(-/-) mice. Moreover, FoxO1, SOD2 and Nrf2 expression levels were reduced and induction of superoxide dismutase (SOD) and glutathione peroxidase activity was not observed in PQ-treated Casp2(-/-) mice. Strong c-Jun amino-terminal kinase (JNK) activation was observed in Casp2(-/-) mice, indicative of increased stress. Together, our data strongly suggest that caspase-2 deficiency leads to increased cellular stress largely because these mice fail to respond to oxidative stress by upregulating their antioxidant defense mechanism. This makes the mice more vulnerable to exogenous challenges and may partly explain the shorter lifespan of Casp2(-/-) mice.


Subject(s)
Caspase 2/metabolism , Oxidative Stress , Animals , Caspase 2/genetics , Herbicides/toxicity , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Mice , Mice, Knockout , Oxidative Stress/drug effects , Paraquat/toxicity
9.
Cell Death Dis ; 5: e1383, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25144718

ABSTRACT

Caspase-2 has been implicated in various cellular functions, including cell death by apoptosis, oxidative stress response, maintenance of genomic stability and tumor suppression. The loss of the caspase-2 gene (Casp2) enhances oncogene-mediated tumorigenesis induced by E1A/Ras in athymic nude mice, and also in the Eµ-Myc lymphoma and MMTV/c-neu mammary tumor mouse models. To further investigate the function of caspase-2 in oncogene-mediated tumorigenesis, we extended our studies in the TH-MYCN transgenic mouse model of neuroblastoma. Surprisingly, we found that loss of caspase-2 delayed tumorigenesis in the TH-MYCN neuroblastoma model. In addition, tumors from TH-MYCN/Casp2(-/-) mice were predominantly thoracic paraspinal tumors and were less vascularized compared with tumors from their TH-MYCN/Casp2(+/+) counterparts. We did not detect any differences in the expression of neuroblastoma-associated genes in TH-MYCN/Casp2(-/-) tumors, or in the activation of Ras/MAPK signaling pathway that is involved in neuroblastoma progression. Analysis of expression array data from human neuroblastoma samples showed a correlation between low caspase-2 levels and increased survival. However, caspase-2 levels correlated with clinical outcome only in the subset of MYCN-non-amplified human neuroblastoma. These observations indicate that caspase-2 is not a suppressor in MYCN-induced neuroblastoma and suggest a tissue and context-specific role for caspase-2 in tumorigenesis.


Subject(s)
Caspase 2/metabolism , Neuroblastoma/pathology , Animals , Caspase 2/deficiency , Caspase 2/genetics , Disease Models, Animal , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma/metabolism , Neuroblastoma/mortality , Signal Transduction , ras Proteins/metabolism
10.
Cell Death Differ ; 21(3): 475-80, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24292555

ABSTRACT

A recent report claimed that endoplasmic reticulum (ER) stress activates the ER trans-membrane receptor IRE1α, leading to increased caspase-2 levels via degradation of microRNAs, and consequently induction of apoptosis. This observation casts caspase-2 into a central role in the apoptosis triggered by ER stress. We have used multiple cell types from caspase-2-deficient mice to test this hypothesis but failed to find significant impact of loss of caspase-2 on ER-stress-induced apoptosis. Moreover, we did not observe increased expression of caspase-2 protein in response to ER stress. Our data strongly argue against a critical role for caspase-2 in ER-stress-induced apoptosis.


Subject(s)
Caspase 2/metabolism , Cysteine Endopeptidases/metabolism , Endoplasmic Reticulum Stress/physiology , Animals , Caspase 2/genetics , Cysteine Endopeptidases/genetics , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thymocytes/enzymology , Thymocytes/metabolism , Up-Regulation , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
11.
Cell Death Differ ; 20(9): 1133-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23811850

ABSTRACT

Ever since its discovery 20 years ago, caspase-2 has been enigmatic and its function somewhat controversial. Although many in vitro studies suggested that caspase-2 was important for apoptosis, demonstrating an in vivo cell death role for this caspase has been more problematic, with caspase-2-deficient mice showing limited, tissue-specific cell death defects. Recent results from different laboratories suggest that at least one of its physiological roles in animals is to protect against cellular stress and transformation. As such, loss of caspase-2 augments tumorigenesis in some mouse models of cancer, assigning a tumour suppressor function to this enigmatic caspase. This review focuses on this seemingly non-apoptotic function of caspase-2 as a tumour suppressor and reconciles some of the recent findings in the field.


Subject(s)
Apoptosis/genetics , Caspase 2/genetics , Caspase 2/metabolism , Cell Transformation, Neoplastic/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle/genetics , DNA Repair/genetics , Genes, Tumor Suppressor , Mice , Mice, Knockout , Neoplasms/genetics , Tumor Suppressor Proteins/metabolism
13.
Cell Death Differ ; 19(8): 1288-98, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22498700

ABSTRACT

Caspase-2 is an initiator caspase, which has been implicated to function in apoptotic and non-apoptotic signalling pathways, including cell-cycle regulation, DNA-damage signalling and tumour suppression. We previously demonstrated that caspase-2 deficiency enhances E1A/Ras oncogene-induced cell transformation and augments lymphomagenesis in the EµMyc mouse model. Caspase-2(-/-) mouse embryonic fibroblasts (casp2(-/-) MEFs) show aberrant cell-cycle checkpoint regulation and a defective apoptotic response following DNA damage. Disruption of cell-cycle checkpoints often leads to genomic instability (GIN), which is a common phenotype of cancer cells and can contribute to cellular transformation. Here we show that caspase-2 deficiency results in increased DNA damage and GIN in proliferating cells. Casp2(-/-) MEFs readily escape senescence in culture and exhibit increased micronuclei formation and sustained DNA damage during cell culture and following γ-irradiation. Metaphase analyses demonstrated that a lack of caspase-2 is associated with increased aneuploidy in both MEFs and in EµMyc lymphoma cells. In addition, casp2(-/-) MEFs and lymphoma cells exhibit significantly decreased telomere length. We also noted that loss of caspase-2 leads to defective p53-mediated signalling and decreased trans-activation of p53 target genes upon DNA damage. Our findings suggest that loss of caspase-2 serves as a key function in maintaining genomic integrity, during cell proliferation and following DNA damage.


Subject(s)
Caspase 2/deficiency , DNA Damage , Genomic Instability , Aneuploidy , Animals , Caspase 2/genetics , Caspase 2/metabolism , Cell Growth Processes/genetics , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mice , Mice, Knockout , Signal Transduction , Transfection
14.
Cell Death Differ ; 19(8): 1370-80, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22343713

ABSTRACT

Caspase-2 has been implicated in apoptosis and in non-apoptotic processes such as cell cycle regulation, tumor suppression and ageing. Using caspase-2 knockout (casp2(-/-)) mice, we show here that the putative anti-ageing role of this caspase is due in part to its involvement in the stress response pathway. The old casp2(-/-) mice show increased cellular levels of oxidized proteins, lipid peroxides and DNA damage, suggesting enhanced oxidative stress. Furthermore, murine embryonic fibroblasts from casp2(-/-) mice showed increased reactive oxygen species generation when challenged with pro-oxidants. Reduced activities of antioxidant enzymes glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) were observed in the old casp2(-/-) mice. Interestingly, in the old casp2(-/-) animals expression of FoxO1 and FoxO3a was significantly reduced, whereas p21 levels and the number of senescent hepatocytes were elevated. In contrast to young wild-type mice, the casp2(-/-) animals fed an on ethanol-based diet failed to show enhanced GSH-Px and SOD activities. Thus, caspase-2, most likely via FoxO transcription factors, regulates the oxidative stress response in vivo.


Subject(s)
Antioxidants/metabolism , Caspase 2/deficiency , Oxidative Stress/physiology , Age Factors , Animals , Apoptosis/physiology , Caspase 2/metabolism , Cellular Senescence/physiology , DNA Damage , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Glutathione Peroxidase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/genetics
15.
Oncogene ; 27(24): 3393-404, 2008 May 29.
Article in English | MEDLINE | ID: mdl-18193089

ABSTRACT

Caspase-2 is one of the most conserved caspases, yet its biological function remains a matter of controversy. In the present article we analysed mouse embryonic fibroblasts (MEFs) from caspase-2 knockout mice for their sensitivity to various apoptosis inducing agents. We found that cell death induced by drugs that disrupt cytoskeleton is significantly inhibited in Casp2(-/-) MEFs. These drugs included zoledronic acid, vincristine, cytochalasin D and paclitaxel. We demonstrate that MEFs lacking Casp2 show clonogenic survival following drug treatment, whereas all Casp2(+/+) MEFs die, indicating that caspase-2 is required for apoptosis induced by cytoskeletal disruption. We further found that caspase-2 mediates apoptosis via Piddosome, Bid and Bax activation, and cytochrome c release. In the absence of caspase-2, Bid and Bax activation, and cytochrome c release are significantly delayed following drug treatment. Our data provide strong support for a context-dependent function of caspase-2 in apoptosis.


Subject(s)
Apoptosis/physiology , Caspase 2/physiology , Cytoskeleton/metabolism , Fibroblasts/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein/metabolism , Bone Density Conservation Agents/pharmacology , CRADD Signaling Adaptor Protein , Carrier Proteins , Cells, Cultured , Colony-Forming Units Assay , Cytochromes c/metabolism , Death Domain Receptor Signaling Adaptor Proteins , Diphosphonates/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Imidazoles/pharmacology , Immunoblotting , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Zoledronic Acid , bcl-2-Associated X Protein/metabolism
16.
Cell Death Differ ; 15(3): 461-70, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18084239

ABSTRACT

The activation of caspases is the principal event in the execution of apoptosis. Initiator caspases are activated through an autocatalytic mechanism often involving dimerisation or oligomerisation. In Drosophila, the only initiator caspase DRONC, is tightly inhibited by DIAP1 and removal of DIAP1 permits activation of DRONC by the Drosophila Apaf-1-related killer, ARK. ARK is proposed to facilitate DRONC oligomerisation and autoprocessing at residue E352. This study examines whether autoprocessing of DRONC is required for its activation and for DRONC-mediated cell death. Using purified recombinant proteins, we show here that while DRONC autocleaves at residue E352, mutation of this site did not abolish enzyme activation, DRICE-induced cleavage of DRONC or DRONC-mediated activation of DRICE. We performed a detailed mutational analysis of DRONC cleavage sites and show that overexpression of DRONC cleavage mutants in Drosophila cells retain pro-apoptotic activity. Using an in vitro cell-free assay, we found ARK alone did not activate DRONC and demonstrate a requirement for an additional cytosolic factor in ARK-mediated DRONC activation. These results suggest that, similar to mammalian caspase-2 and caspase-9, the initial cleavage of DRONC is not essential for its activation and suggest a mechanism of ARK-mediated DRONC activation different from that proposed previously.


Subject(s)
Apoptosis , Caspases/metabolism , Drosophila Proteins/metabolism , Drosophila/enzymology , Enzyme Precursors/metabolism , Animals , Aspartic Acid/analysis , Caspases/chemistry , Caspases/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Enzyme Activation , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Glutamic Acid/analysis , Inhibitor of Apoptosis Proteins/metabolism , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
17.
Cell Death Differ ; 14(5): 907-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17205077

ABSTRACT

The Bcl-2-family of proteins localize to intracellular membranes via a C-terminal hydrophobic membrane anchor (MA) domain, to exert their antiapoptotic or proapoptotic functions. In Drosophila, both Bcl-2 family members, DEBCL and BUFFY, contain an MA. In DEBCL the MA is necessary for the localization of protein to mitochondria and for its proapoptotic activity. BUFFY is highly similar to DEBCL but its localization and function are not clearly defined. Here, we report on comparative analysis of BUFFY and DEBCL to decipher the molecular basis for their subcellular localization. We show that these two proteins localize to distinct intracellular membranes, DEBCL predominantly to mitochondria and BUFFY to endoplasmic reticula (ER). Our results suggest that the MA-flanking residues in DEBCL, homologous to Bcl-X(L), are required for the targeting of DEBCL to mitochondria. The C-terminal positively charged residues present in DEBCL are absent in BUFFY, which allows for its localization to ER. The MA in both proteins is required for the correct targeting and proapoptotic activities of these proteins. Interestingly, a functional nuclear localization signal was identified in the N-terminal region of BUFFY and in the absence of the MA, BUFFY accumulated in the nucleus. The functional implications of these findings are discussed.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Apoptosis , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Drosophila Proteins/chemistry , Endoplasmic Reticulum/metabolism , Humans , Membrane Proteins/chemistry , Mice , Mitochondrial Membranes/metabolism , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , NIH 3T3 Cells , Nuclear Localization Signals/metabolism , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins c-bcl-2/metabolism , Subcellular Fractions/metabolism , bcl-X Protein/chemistry , bcl-X Protein/metabolism
19.
J Biol Chem ; 276(27): 25342-50, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11337486

ABSTRACT

Caspases are main effectors of apoptosis in metazoans. Genome analysis indicates that there are seven caspases in Drosophila, six of which have been previously characterized. Here we describe the cloning and characterization of the last Drosophila caspase, DAMM. Similar to mammalian effector caspases, DAMM lacks a long prodomain. We show that the DAMM precursor, along with the caspases DRONC and DECAY, is partially processed in cells undergoing apoptosis. Recombinant DAMM produced in Escherichia coli shows significant catalytic activity on a pentapeptide caspase substrate. Low levels of damm mRNA are ubiquitously expressed in Drosophila embryos during early stages of development. Relatively high levels of damm mRNA are detected in larval salivary glands and midgut, and in adult egg chambers. Ectopic expression of DAMM in cultured cells induces apoptosis, and similarly, transgenic overexpression of DAMM, but not of a catalytically inactive DAMM mutant, in Drosophila results in a rough eye phenotype. We demonstrate that expression of the catalytically inactive DAMM mutant protein significantly suppresses the rough eye phenotype due to the overexpression of HID, suggesting that DAMM may be required in a hid-mediated cell death pathway.


Subject(s)
Caspases/chemistry , Drosophila Proteins , Drosophila/enzymology , Amino Acid Sequence , Animals , Apoptosis , Caspases/genetics , Caspases/metabolism , Catalysis , Cells, Cultured , Cloning, Molecular , Drosophila/growth & development , Escherichia coli , Eye/growth & development , Inhibitor of Apoptosis Proteins , Insect Proteins/metabolism , Molecular Sequence Data , Phenotype , RNA, Messenger/metabolism , Recombinant Proteins/metabolism
20.
J Biol Chem ; 275(51): 40416-24, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-10984473

ABSTRACT

Dronc is a caspase recruitment domain-containing Drosophila caspase that is expressed in a temporally and spatially restricted fashion during development. Dronc is the only fly caspase known to be regulated by the hormone ecdysone. Here we show that ectopic expression of dronc in the developing fly eye leads to increased cell death and an ablated eye phenotype that can be suppressed by halving the dosage of the genes in the H99 complex (reaper, hid, and grim) and enhanced by mutations in diap1. In contrast to previous reports, we show that the dronc eye ablation phenotype can be suppressed by coexpression of the baculoviral caspase inhibitor p35. Dronc also interacts, both genetically and biochemically, with the CED-4/Apaf-1 fly homolog, Dark. Furthermore, extracts made from Dark homozygous mutant flies have reduced ability to process Dronc, showing that Dark is required for Dronc processing. Finally, using the RNA interference technique, we show that loss of Dronc function in early Drosophila embryos results in a dramatic decrease in cell death, indicating that Dronc is important for programmed cell death during embryogenesis. These results suggest that Dronc is a key caspase mediating programmed cell death in Drosophila.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Drosophila Proteins , Drosophila/cytology , Animals , Animals, Genetically Modified , Caspases/genetics , Darkness , Embryo, Nonmammalian/cytology , Eye/metabolism , Phenotype , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viral Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...