Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
FEBS J ; 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38069549

ABSTRACT

Cells respond to invading pathogens and danger signals from the environment by adapting gene expression to meet the need for protective effector molecules. While this innate immune response is required for the cell and the organism to recover, excess immune activation may lead to loss of homeostasis, thereby promoting chronic inflammation and cancer progression. The molecular basis of innate immune defence is comprised of factors promoting survival and proliferation, such as cytokines, antimicrobial peptides and anti-apoptotic proteins. As the molecular mechanisms regulating innate immune responses are conserved through evolution, the fruit fly Drosophila melanogaster serves as a convenient, affordable and ethical model organism to enhance understanding of immune signalling. Fly immunity against bacterial infection is built up by both cellular and humoral responses, where the latter is regulated by the Imd and Toll pathways activating NF-κB transcription factors Relish, Dorsal and Dif, as well as JNK activation and JAK/STAT signalling. As in mammals, the Drosophila innate immune signalling pathways are characterised by ubiquitination of signalling molecules followed by ubiquitin receptors binding to the ubiquitin chains, as well as by rapid changes in protein levels by ubiquitin-mediated targeted proteasomal and lysosomal degradation. In this review, we summarise the molecular signalling pathways regulating immune responses to pathogen infection in Drosophila, with a focus on ubiquitin-dependent control of innate immunity and inflammatory signalling.

2.
Front Immunol ; 14: 1253805, 2023.
Article in English | MEDLINE | ID: mdl-37809071

ABSTRACT

Introduction: Stilbenoid compounds have been described to have anti-inflammatory properties in animal models in vivo, and have been shown to inhibit Ca2+-influx through the transient receptor potential ankyrin 1 (TrpA1). Methods: To study how stilbenoid compounds affect inflammatory signaling in vivo, we have utilized the fruit fly, Drosophila melanogaster, as a model system. To induce intestinal inflammation in the fly, we have fed flies with the intestinal irritant dextran sodium sulphate (DSS). Results: We found that DSS induces severe changes in the bacteriome of the Drosophila intestine, and that this dysbiosis causes activation of the NF-κB transcription factor Relish. We have taken advantage of the DSS-model to study the anti-inflammatory properties of the stilbenoid compounds pinosylvin (PS) and pinosylvin monomethyl ether (PSMME). With the help of in vivo approaches, we have identified PS and PSMME to be transient receptor ankyrin 1 (TrpA1)-dependent antagonists of NF-κB-mediated intestinal immune responses in Drosophila. We have also computationally predicted the putative antagonist binding sites of these compounds at Drosophila TrpA1. Discussion: Taken together, we show that the stilbenoids PS and PSMME have anti-inflammatory properties in vivo in the intestine and can be used to alleviate chemically induced intestinal inflammation in Drosophila.


Subject(s)
NF-kappa B , Stilbenes , Animals , NF-kappa B/metabolism , Drosophila/metabolism , Drosophila melanogaster/metabolism , Ankyrins , Intestines , Stilbenes/pharmacology , Anti-Inflammatory Agents/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy
3.
J Cell Sci ; 136(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36695453

ABSTRACT

The heat shock (HS) response is crucial for cell survival in harmful environments. Nuclear lamin A/C, encoded by the LMNA gene, contributes towards altered gene expression during HS, but the underlying mechanisms are poorly understood. Here, we show that upon HS, lamin A/C was reversibly phosphorylated at serine 22 in concert with HSF1 activation in human cells, mouse cells and Drosophila melanogaster in vivo. Consequently, the phosphorylation facilitated nucleoplasmic localization of lamin A/C and nuclear sphericity in response to HS. Interestingly, lamin A/C knock-out cells showed deformed nuclei after HS and were rescued by ectopic expression of wild-type lamin A, but not by a phosphomimetic (S22D) lamin A mutant. Furthermore, HS triggered concurrent downregulation of lamina-associated protein 2α (Lap2α, encoded by TMPO) in wild-type lamin A/C-expressing cells, but a similar response was perturbed in lamin A/C knock-out cells and in LMNA mutant patient fibroblasts, which showed impaired cell cycle arrest under HS and compromised survival at recovery. Taken together, our results suggest that the altered phosphorylation stoichiometry of lamin A/C provides an evolutionarily conserved mechanism to regulate lamina structure and serve nuclear adaptation and cell survival during HS.


Subject(s)
Lamin Type A , Serine , Humans , Mice , Animals , Lamin Type A/genetics , Phosphorylation , Serine/metabolism , Drosophila melanogaster/metabolism , Cell Nucleus/metabolism
4.
Cell Death Differ ; 30(2): 227-236, 2023 02.
Article in English | MEDLINE | ID: mdl-35810247

ABSTRACT

An intact cell death machinery is not only crucial for successful embryonic development and tissue homeostasis, but participates also in the defence against pathogens and contributes to a balanced immune response. Centrally involved in the regulation of both cell death and inflammatory immune responses is the evolutionarily conserved family of cysteine proteases named caspases. The Drosophila melanogaster genome encodes for seven caspases, several of which display dual functions, participating in apoptotic signalling and beyond. Among the Drosophila caspases, the caspase-8 homologue Dredd has a well-characterised role in inflammatory signalling activated by bacterial infections, and functions as a driver of NF-κB-mediated immune responses. Regarding the other Drosophila caspases, studies focusing on tissue-specific immune signalling and host-microbe interactions have recently revealed immunoregulatory functions of the initiator caspase Dronc and the effector caspase Drice. The aim of this review is to give an overview of the signalling cascades involved in the Drosophila humoral innate immune response against pathogens and of their caspase-mediated regulation. Furthermore, the apoptotic role of caspases during antibacterial and antiviral immune activation will be discussed.


Subject(s)
Caspases , Drosophila Proteins , Animals , Caspases/genetics , Caspases/metabolism , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Host Microbial Interactions , Apoptosis
5.
FEBS J ; 290(9): 2489-2502, 2023 05.
Article in English | MEDLINE | ID: mdl-36239424

ABSTRACT

Lactate dehydrogenase A (LDHA) is a glycolytic enzyme catalysing the reversible conversion of pyruvate to lactate. It has been implicated as a substrate for PIM kinases, yet the relevant target sites and functional consequences of phosphorylation have remained unknown. Here, we show that all three PIM family members can phosphorylate LDHA at serine 161. When we investigated the physiological consequences of this phosphorylation in PC3 prostate cancer and MCF7 breast cancer cells, we noticed that it suppressed ubiquitin-mediated degradation of nuclear LDHA and promoted interactions between LDHA and 14-3-3 proteins. By contrast, in CRISPR/Cas9-edited knock-out cells lacking all three PIM family members, ubiquitination of nuclear LDHA was dramatically increased followed by its decreased expression. Our data suggest that PIM kinases support nuclear LDHA expression and activities by promoting phosphorylation-dependent interactions of LDHA with 14-3-3ε, which shields nuclear LDHA from ubiquitin-mediated degradation.


Subject(s)
Lactate Dehydrogenase 5 , Protein Serine-Threonine Kinases , Serine , Humans , Cell Line, Tumor , Lactate Dehydrogenase 5/metabolism , Serine/genetics , Serine/metabolism , Ubiquitination , Ubiquitins/metabolism , Protein Serine-Threonine Kinases/metabolism
6.
Front Cell Dev Biol ; 10: 932983, 2022.
Article in English | MEDLINE | ID: mdl-36111332

ABSTRACT

Lamins A and C are nuclear intermediate filament proteins that form a proteinaceous meshwork called lamina beneath the inner nuclear membrane. Mutations in the LMNA gene encoding lamins A and C cause a heterogenous group of inherited degenerative diseases known as laminopathies. Previous studies have revealed altered cell signaling pathways in lamin-mutant patient cells, but little is known about the fate of mutant lamins A and C within the cells. Here, we analyzed the turnover of lamins A and C in cells derived from a dilated cardiomyopathy patient with a heterozygous p.S143P mutation in LMNA. We found that transcriptional activation and mRNA levels of LMNA are increased in the primary patient fibroblasts, but the protein levels of lamins A and C remain equal in control and patient cells because of a meticulous interplay between autophagy and the ubiquitin-proteasome system (UPS). Both endogenous and ectopic expression of p.S143P lamins A and C cause significantly reduced activity of UPS and an accumulation of K48-ubiquitin chains in the nucleus. Furthermore, K48-ubiquitinated lamins A and C are degraded by compensatory enhanced autophagy, as shown by increased autophagosome formation and binding of lamins A and C to microtubule-associated protein 1A/1B-light chain 3. Finally, chaperone 4-PBA augmented protein degradation by restoring UPS activity as well as autophagy in the patient cells. In summary, our results suggest that the p.S143P-mutant lamins A and C have overloading and deleterious effects on protein degradation machinery and pharmacological interventions with compounds enhancing protein degradation may be beneficial for cell homeostasis.

7.
Biomater Adv ; 133: 112607, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35525761

ABSTRACT

Due to its modular and flexible design options, mesoporous silica provides ample opportunities when developing new strategies for combinatory antibacterial treatments. In this study, antibacterial ceria (CeO2) nanoparticles (NP) were used as core material, and were further coated with a mesoporous silica shell (mSiO2) to obtain a core@shell structured nanocomposite (CeO2@mSiO2). The porous silica shell was utilized as drug reservoir, whereby CeO2@mSiO2 was loaded with the antimicrobial agent capsaicin (CeO2@mSiO2/Cap). CeO2@mSiO2/Cap was further surface-coated with the natural antimicrobial polymer chitosan by employing physical adsorption. The obtained nanocomposite, CeO2@mSiO2/Cap@Chit, denoted NAB, which stands for "nanoantibiotic", provided a combinatory antibacterial mode of action. The antibacterial effect of NAB on the Gram-negative bacteria Escherichia coli (E.coli) was proven to be significant in vitro. In addition, in vivo evaluations revealed NAB to inhibit the bacterial growth in the intestine of bacteria-fed Drosophila melanogaster larvae, and decreased the required dose of capsaicin needed to eliminate bacteria. As our constructed CeO2@mSiO2 did not show toxicity to mammalian cells, it holds promise for the development of next-generation nanoantibiotics of non-toxic nature with flexible design options.


Subject(s)
Nanoparticles , Silicon Dioxide , Animals , Anti-Bacterial Agents/pharmacology , Capsaicin , Drosophila melanogaster , Mammals , Nanoparticles/therapeutic use , Silicon Dioxide/pharmacology
8.
FEBS J ; 289(17): 5180-5197, 2022 09.
Article in English | MEDLINE | ID: mdl-35263507

ABSTRACT

Methionine 1 (M1)-linked ubiquitination plays a key role in the regulation of inflammatory nuclear factor-κB (NF-κB) signalling and is important for clearance of pathogen infection in Drosophila melanogaster. M1-linked ubiquitin (M1-Ub) chains are assembled by the linear ubiquitin E3 ligase (LUBEL) in flies. Here, we have studied the role of LUBEL in sterile inflammation induced by different types of cellular stresses. We have found that the LUBEL catalyses formation of M1-Ub chains in response to hypoxic, oxidative and mechanical stress conditions. LUBEL is shown to be important for flies to survive low oxygen conditions and paraquat-induced oxidative stress. This protective action seems to be driven by stress-induced activation of the NF-κB transcription factor Relish via the immune deficiency (Imd) pathway. In addition to LUBEL, the intracellular mediators of Relish activation, including the transforming growth factor activating kinase 1 (Tak1), Drosophila inhibitor of apoptosis (IAP) Diap2, the IκB kinase γ (IKKγ) Kenny and the initiator caspase Death-related ced-3/Nedd2-like protein (Dredd), but not the membrane receptor peptidoglycan recognition protein (PGRP)-LC, are shown to be required for sterile inflammatory response and survival. Finally, we showed that the stress-induced upregulation of M1-Ub chains in response to hypoxia, oxidative and mechanical stress is also induced in mammalian cells and protects from stress-induced cell death. Taken together, our results suggest that M1-Ub chains are important for NF-κB signalling in inflammation induced by stress conditions often observed in chronic inflammatory diseases and cancer.


Subject(s)
Drosophila Proteins , NF-kappa B , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Inflammation/genetics , MAP Kinase Kinase Kinases/metabolism , Mammals/metabolism , Methionine/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Ubiquitin/metabolism , Ubiquitination
9.
Cell Death Differ ; 29(1): 28-39, 2022 01.
Article in English | MEDLINE | ID: mdl-34262145

ABSTRACT

The Drosophila IAP protein, Diap2, is a key mediator of NF-κB signalling and innate immune responses. Diap2 is required for both local immune activation, taking place in the epithelial cells of the gut and trachea, and for mounting systemic immune responses in the cells of the fat body. We have found that transgenic expression of Diap2 leads to a spontaneous induction of NF-κB target genes, inducing chronic inflammation in the Drosophila midgut, but not in the fat body. Drice is a Drosophila effector caspase known to interact and form a stable complex with Diap2. We have found that this complex formation induces its subsequent degradation, thereby regulating the amount of Diap2 driving NF-κB signalling in the intestine. Concordantly, loss of Drice activity leads to accumulation of Diap2 and to chronic intestinal inflammation. Interestingly, Drice does not interfere with pathogen-induced signalling, suggesting that it protects from immune responses induced by resident microbes. Accordingly, no inflammation was detected in transgenic Diap2 flies and Drice-mutant flies reared in axenic conditions. Hence, we show that Drice, by restraining Diap2, halts unwanted inflammatory signalling in the intestine.


Subject(s)
Drosophila Proteins/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Signal Transduction , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Immunity, Innate , Inflammation
10.
Cancer Res ; 80(7): 1414-1427, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32029551

ABSTRACT

For maximal oncogenic activity, cellular MYC protein levels need to be tightly controlled so that they do not induce apoptosis. Here, we show how ubiquitin ligase UBR5 functions as a molecular rheostat to prevent excess accumulation of MYC protein. UBR5 ubiquitinates MYC and its effects on MYC protein stability are independent of FBXW7. Silencing of endogenous UBR5 induced MYC protein expression and regulated MYC target genes. Consistent with the tumor suppressor function of UBR5 (HYD) in Drosophila, HYD suppressed dMYC-dependent overgrowth of wing imaginal discs. In contrast, in cancer cells, UBR5 suppressed MYC-dependent priming to therapy-induced apoptosis. Of direct cancer relevance, MYC and UBR5 genes were coamplified in MYC-driven human cancers. Functionally, UBR5 suppressed MYC-mediated apoptosis in p53-mutant breast cancer cells with UBR5/MYC coamplification. Furthermore, single-cell immunofluorescence analysis demonstrated reciprocal expression of UBR5 and MYC in human basal-type breast cancer tissues. In summary, UBR5 is a novel MYC ubiquitin ligase and an endogenous rheostat for MYC activity. In MYC-amplified, and p53-mutant breast cancer cells, UBR5 has an important role in suppressing MYC-mediated apoptosis priming and in protection from drug-induced apoptosis. SIGNIFICANCE: These findings identify UBR5 as a novel MYC regulator, the inactivation of which could be very important for understanding of MYC dysregulation on cancer cells. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/7/1414/F1.large.jpg.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Animals, Genetically Modified , Apoptosis/genetics , Breast/pathology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Models, Animal , Protein Stability , Proto-Oncogene Proteins c-myc/metabolism , RNA-Seq , Tissue Array Analysis , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics
11.
Cell Death Differ ; 26(5): 860-876, 2019 05.
Article in English | MEDLINE | ID: mdl-30026495

ABSTRACT

Post-translational modifications such as ubiquitination play a key role in regulation of inflammatory nuclear factor-κB (NF-κB) signalling. The Drosophila IκB kinase γ (IKKγ) Kenny is a central regulator of the Drosophila Imd pathway responsible for activation of the NF-κB Relish. We found the Drosophila E3 ligase and HOIL-1L interacting protein (HOIP) orthologue linear ubiquitin E3 ligase (LUBEL) to catalyse formation of M1-linked linear ubiquitin (M1-Ub) chains in flies in a signal-dependent manner upon bacterial infection. Upon activation of the Imd pathway, LUBEL modifies Kenny with M1-Ub chains. Interestingly, the LUBEL-mediated M1-Ub chains seem to be targeted both directly to Kenny and to K63-linked ubiquitin chains conjugated to Kenny by DIAP2. This suggests that DIAP2 and LUBEL work together to promote Kenny-mediated activation of Relish. We found LUBEL-mediated M1-Ub chain formation to be required for flies to survive oral infection with Gram-negative bacteria, for activation of Relish-mediated expression of antimicrobial peptide genes and for pathogen clearance during oral infection. Interestingly, LUBEL is not required for mounting an immune response against systemic infection, as Relish-mediated antimicrobial peptide genes can be expressed in the absence of LUBEL during septic injury. Finally, transgenic induction of LUBEL-mediated M1-Ub drives expression of antimicrobial peptide genes and hyperplasia in the midgut in the absence of infection. This suggests that M1-Ub chains are important for Imd signalling and immune responses in the intestinal epithelia, and that enhanced M1-Ub chain formation is able to drive chronic intestinal inflammation in flies.


Subject(s)
Bacterial Infections/genetics , Drosophila Proteins/genetics , Inflammation/genetics , Inhibitor of Apoptosis Proteins/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Bacterial Infections/microbiology , Disease Models, Animal , Drosophila/genetics , Gram-Negative Bacteria/pathogenicity , Humans , Immunity, Innate/genetics , Inflammation/microbiology , Mouth/microbiology , Mouth/pathology , NF-kappa B/genetics , Protein Processing, Post-Translational/genetics , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Ubiquitin/genetics , Ubiquitination/genetics
12.
Curr Protoc Toxicol ; 77(1): e52, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29933523

ABSTRACT

As several diseases have been linked to dysbiosis of the human intestinal microflora, manipulation of the microbiota has emerged as an exciting new strategy for potentially treating and preventing diseases. However, the human microbiota consists of a plethora of different species, and distinguishing the impact of a specific bacterial species on human health is challenging. In tackling this challenge, the fruit fly Drosophila melanogaster, with its far simpler microbial composition, has emerged as a powerful model for unraveling host-microbe interactions. To study the interplay between the resident commensal microbiome and the host, flies can be made germ-free, or axenic. To elucidate the impact of specific bacteria, axenic flies can then be re-introduced to specific microbial species. In this unit, we provide a step-by-step protocol on how to rear Drosophila melanogaster under axenic conditions and confirm the axenity of flies. © 2018 by John Wiley & Sons, Inc.

13.
Adv Healthc Mater ; 6(21)2017 Nov.
Article in English | MEDLINE | ID: mdl-28892296

ABSTRACT

Approaches to increase the efficiency in developing drugs and diagnostics tools, including new drug delivery and diagnostic technologies, are needed for improved diagnosis and treatment of major diseases and health problems such as cancer, inflammatory diseases, chronic wounds, and antibiotic resistance. Development within several areas of research ranging from computational sciences, material sciences, bioengineering to biomedical sciences and bioimaging is needed to realize innovative drug development and diagnostic (DDD) approaches. Here, an overview of recent progresses within key areas that can provide customizable solutions to improve processes and the approaches taken within DDD is provided. Due to the broadness of the area, unfortunately all relevant aspects such as pharmacokinetics of bioactive molecules and delivery systems cannot be covered. Tailored approaches within (i) bioinformatics and computer-aided drug design, (ii) nanotechnology, (iii) novel materials and technologies for drug delivery and diagnostic systems, and (iv) disease models to predict safety and efficacy of medicines under development are focused on. Current developments and challenges ahead are discussed. The broad scope reflects the multidisciplinary nature of the field of DDD and aims to highlight the convergence of biological, pharmaceutical, and medical disciplines needed to meet the societal challenges of the 21st century.


Subject(s)
Models, Biological , Computational Biology , Drug Delivery Systems , Drug Design , Humans , Molecular Docking Simulation , Nanomedicine , Nanotechnology , Neoplasms/diagnosis , Proteins/chemistry , Proteins/metabolism , Quantitative Structure-Activity Relationship
14.
FASEB J ; 31(12): 5332-5341, 2017 12.
Article in English | MEDLINE | ID: mdl-28778974

ABSTRACT

Cytoplasmic intermediate filaments (cIFs) are found in all eumetazoans, except arthropods. To investigate the compatibility of cIFs in arthropods, we expressed human vimentin (hVim), a cIF with filament-forming capacity in vertebrate cells and tissues, transgenically in Drosophila Transgenic hVim could be recovered from whole-fly lysates by using a standard procedure for intermediate filament (IF) extraction. When this procedure was used to test for the possible presence of IF-like proteins in flies, only lamins and tropomyosin were observed in IF-enriched extracts, thereby providing biochemical reinforcement to the paradigm that arthropods lack cIFs. In Drosophila, transgenic hVim was unable to form filament networks in S2 cells and mesenchymal tissues; however, cage-like vimentin structures could be observed around the nuclei in internal epithelia, which suggests that Drosophila retains selective competence for filament formation. Taken together, our results imply that although the filament network formation competence is partially lost in Drosophila, a rudimentary filament network formation ability remains in epithelial cells. As a result of the observed selective competence for cIF assembly in Drosophila, we hypothesize that internal epithelial cIFs were the last cIFs to disappear from arthropods.-Gullmets, J., Torvaldson, E., Lindqvist, J., Imanishi, S. Y., Taimen, P., Meinander, A., Eriksson, J. E. Internal epithelia in Drosophila display rudimentary competence to form cytoplasmic networks of transgenic human vimentin.


Subject(s)
Cytoplasm/metabolism , Drosophila/metabolism , Epithelium/metabolism , Vimentin/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Intermediate Filaments/enzymology , Intermediate Filaments/metabolism , Lamins/genetics , Lamins/metabolism , Tropomyosin/genetics , Tropomyosin/metabolism , Vimentin/genetics
15.
EMBO J ; 31(12): 2770-83, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22549468

ABSTRACT

Caspases have been extensively studied as critical initiators and executioners of cell death pathways. However, caspases also take part in non-apoptotic signalling events such as the regulation of innate immunity and activation of nuclear factor-κB (NF-κB). How caspases are activated under these conditions and process a selective set of substrates to allow NF-κB signalling without killing the cell remains largely unknown. Here, we show that stimulation of the Drosophila pattern recognition protein PGRP-LCx induces DIAP2-dependent polyubiquitylation of the initiator caspase DREDD. Signal-dependent ubiquitylation of DREDD is required for full processing of IMD, NF-κB/Relish and expression of antimicrobial peptide genes in response to infection with Gram-negative bacteria. Our results identify a mechanism that positively controls NF-κB signalling via ubiquitin-mediated activation of DREDD. The direct involvement of ubiquitylation in caspase activation represents a novel mechanism for non-apoptotic caspase-mediated signalling.


Subject(s)
Carrier Proteins/metabolism , Caspases/metabolism , Drosophila Proteins/metabolism , Drosophila/immunology , Gene Expression Regulation , Gram-Negative Bacteria/immunology , Inhibitor of Apoptosis Proteins/metabolism , Ubiquitination , Animals , Antimicrobial Cationic Peptides/biosynthesis , Drosophila/genetics , Drosophila/microbiology , Immunity, Innate , Models, Biological , NF-kappa B/metabolism , Transcription Factors/metabolism
16.
J Biol Chem ; 286(21): 18375-82, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21324892

ABSTRACT

The expression levels of caspase-8 inhibitory c-FLIP proteins play an important role in regulating death receptor-mediated apoptosis, as their concentration at the moment when the death-inducing signaling complex (DISC) is formed determines the outcome of the DISC signal. Experimental studies have shown that c-FLIP proteins are subject to dynamic turnover and that their stability and expression levels can be rapidly altered. Even though the influence of c-FLIP on the apoptotic behavior of a single cell has been captured in mathematical simulation studies, the effect of c-FLIP turnover and stability has not been investigated. In this study, a mathematical model of apoptosis was developed to analyze how the dynamic turnover and stability of the c-FLIP isoforms regulate apoptotic signaling for both individual cells and cell populations. Intercellular parameter and concentration distributions were used to describe the behavior of cell populations. Monte-Carlo simulations of cell populations showed that c-FLIP turnover is a key determinant of death receptor responses. The fact that the developed model simulates the state of whole cell populations makes it possible to validate it by comparison with empirical data. The proposed modeling approach can be used to further determine limiting factors in the DISC signaling process.


Subject(s)
Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Communication/physiology , Models, Biological , Signal Transduction/physiology , fas Receptor/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line, Tumor , Humans , Monte Carlo Method , fas Receptor/genetics
17.
BMC Cell Biol ; 11: 45, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20573281

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate (S1P) is a bioactive lipid that regulates a multitude of cellular functions, including cell proliferation, survival, migration and angiogenesis. S1P mediates its effects either by signaling through G protein-coupled receptors (GPCRs) or through an intracellular mode of action. In this study, we have investigated the mechanism behind S1P-induced survival signalling. RESULTS: We found that S1P protected cells from FasL-induced cell death in an NF-kappaB dependent manner. NF-kappaB was activated by extracellular S1P via S1P2 receptors and Gi protein signaling. Our study also demonstrates that extracellular S1P stimulates cells to rapidly produce and secrete additional S1P, which can further amplify the NF-kappaB activation. CONCLUSIONS: We propose a self-amplifying loop of autocrine S1P with capacity to enhance cell survival. The mechanism provides increased understanding of the multifaceted roles of S1P in regulating cell fate during normal development and carcinogenesis.


Subject(s)
Lysophospholipids/biosynthesis , NF-kappa B/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autocrine Communication , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fas Ligand Protein/metabolism , Feedback, Physiological , Flavonoids/pharmacology , Humans , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/antagonists & inhibitors , Sphingosine/biosynthesis , Sphingosine/genetics , Transgenes/genetics
18.
ACS Nano ; 3(1): 197-206, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19206267

ABSTRACT

Mesoporous silica nanoparticles functionalized by surface hyperbranching polymerization of poly(ethylene imine), PEI, were further modified by introducing both fluorescent and targeting moieties, with the aim of specifically targeting cancer cells. Owing to the high abundance of folate receptors in many cancer cells as compared to normal cells, folic acid was used as the targeting ligand. The internalization of the particles in cell lines expressing different levels of folate receptors was studied. Flow cytometry was used to quantify the mean number of nanoparticles internalized per cell. Five times more particles were internalized by cancer cells expressing folate receptors as compared to the normal cells expressing low levels of the receptor. Not only the number of nanoparticles internalized per cell, but also the fraction of cells that had internalized nanoparticles was higher. The total number of particles internalized by the cancer cells was, therefore, about an order of magnitude higher than the total number of particles internalized by normal cells, a difference high enough to be of significant biological importance. In addition, the biospecifically tagged hybrid PEI-silica particles were shown to be noncytotoxic and able to specifically target folate receptor-expressing cancer cells also under coculture conditions.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Silicon Dioxide/chemistry , Carrier Proteins/chemistry , Coculture Techniques , Flow Cytometry/methods , Folate Receptors, GPI-Anchored , Folic Acid/chemistry , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Nanoparticles/chemistry , Nanotechnology/methods , Neoplasms/pathology , Polyethyleneimine/chemistry , Polymers/chemistry , Porosity , Receptors, Cell Surface/chemistry
19.
Toxicol Appl Pharmacol ; 232(2): 258-67, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18687350

ABSTRACT

Oxidative glutamate toxicity in HT22 murine hippocampal cells is a model for neuronal death by oxidative stress. We have investigated the role of proteases in HT22 cell oxidative glutamate toxicity. L-glutamate-induced toxicity was characterized by cell and nuclear shrinkage and chromatin condensation, yet occurred in the absence of either DNA fragmentation or mitochondrial cytochrome c release. Pretreatment with the selective caspase inhibitors either benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (pan-caspase), N-acetyl-Leu-Glu-His-Asp-aldehyde (caspase 9) or N-acetyl-Ile-Glu-Thr-Asp-aldehyde (caspase 8), significantly increased L-glutamate-induced cell death with a corresponding increase in observed nuclear shrinkage and chromatin condensation. This enhancement of glutamate toxicity correlated with an increase in L-glutamate-dependent production of reactive oxygen species (ROS) as a result of caspase inhibition. Pretreating the cells with N-acetyl-L-cysteine prevented ROS production, cell shrinkage and cell death from L-glutamate as well as that associated with the presence of the pan-caspase inhibitor. In contrast, the caspase-3/-7 inhibitor N-acetyl-Asp-Glu-Val-Asp aldehyde was without significant effect. However, pretreating the cells with the calpain inhibitor N-acetyl-Leu-Leu-Nle-CHO, but not the cathepsin B inhibitor CA-074, prevented cell death. The cytotoxic role of calpains was confirmed further by: 1) cytotoxic dependency on intracellular Ca(2+) increase, 2) increased cleavage of the calpain substrate Suc-Leu-Leu-Val-Tyr-AMC and 3) immunoblot detection of the calpain-selective 145 kDa alpha-fodrin cleavage fragment. We conclude that oxidative L-glutamate toxicity in HT22 cells is mediated via calpain activation, whereas inhibition of caspases-8 and -9 may exacerbate L-glutamate-induced oxidative neuronal damage through increased oxidative stress.


Subject(s)
Calpain/physiology , Caspases/physiology , Glutamic Acid/toxicity , Neurons/enzymology , Oxidative Stress/physiology , Animals , Calpain/antagonists & inhibitors , Caspase Inhibitors , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Mice , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Protease Inhibitors/pharmacology
20.
J Immunol ; 178(6): 3944-53, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339495

ABSTRACT

Fever has a major impact on immune responses by modulating survival, proliferation, and endurance of lymphocytes. Lymphocyte persistence in turn is determined by the equilibrium between death and survival-promoting factors that regulate death receptor signaling in these cells. A potential integrator of death receptor signaling is the caspase-8 inhibitor c-FLIP, the expression of which is dynamically regulated, either rapidly induced or down-regulated. In this study, we show in activated primary human T lymphocytes that hyperthermia corresponding to fever triggered down-regulation of both c-FLIP-splicing variants, c-FLIPshort (c-FLIP(S)) and c-FLIPlong, with consequent sensitization to apoptosis mediated by CD95 (Fas/APO-1). The c-FLIP down-regulation and subsequent sensitization was specific for hyperthermic stress. Additionally, we show that the hyperthermia-mediated down-regulation was due to increased ubiquitination and proteasomal degradation of c-FLIP(S), the stability of which we have shown to be regulated by its C-terminal splicing tail. Furthermore, the induced sensitivity to CD95 ligation was independent of heat shock protein 70, as thermotolerant cells, expressing substantially elevated levels of heat shock protein 70, were not rescued from the effect of hyperthermia-mediated c-FLIP down-regulation. Our findings indicate that fever significantly influences the rate of lymphocyte elimination through depletion of c-FLIP(S). Such a general regulatory mechanism for lymphocyte removal has broad ramifications for fever-mediated regulation of immune responses.


Subject(s)
Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Fever/metabolism , Heat-Shock Response , Protease Inhibitors/metabolism , T-Lymphocytes/metabolism , Alternative Splicing/immunology , Apoptosis/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , Caspase 8/metabolism , Caspase Inhibitors , Down-Regulation , Fever/immunology , Fever/pathology , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/immunology , Heat-Shock Response/immunology , Humans , Jurkat Cells , Protease Inhibitors/immunology , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Ubiquitin/immunology , Ubiquitin/metabolism , fas Receptor/immunology , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...