Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Dev Cell ; 52(3): 335-349.e7, 2020 02 10.
Article En | MEDLINE | ID: mdl-31983631

E2F transcription factors control the cytokinesis machinery and thereby ploidy in hepatocytes. If or how these proteins limit proliferation of polyploid cells with extra centrosomes remains unknown. Here, we show that the PIDDosome, a signaling platform essential for caspase-2-activation, limits hepatocyte ploidy and is instructed by the E2F network to control p53 in the developing as well as regenerating liver. Casp2 and Pidd1 act as direct transcriptional targets of E2F1 and its antagonists, E2F7 and E2F8, that together co-regulate PIDDosome expression during juvenile liver growth and regeneration. Of note, whereas hepatocyte aneuploidy correlates with the basal ploidy state, the degree of aneuploidy itself is not limited by PIDDosome-dependent p53 activation. Finally, we provide evidence that the same signaling network is engaged to control ploidy in the human liver after resection. Our study defines the PIDDosome as a primary target to manipulate hepatocyte ploidy and proliferation rates in the regenerating liver.


Caspase 2/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , E2F Transcription Factors/physiology , Hepatocytes/cytology , Liver Regeneration , Polyploidy , Tumor Suppressor Protein p53/physiology , Aneuploidy , Animals , CRADD Signaling Adaptor Protein/physiology , Centrosome , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cytokinesis , Female , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Knockout
2.
Sci Rep ; 9(1): 3597, 2019 03 05.
Article En | MEDLINE | ID: mdl-30837651

Regulation of lipid absorption by enterocytes can influence metabolic status in humans and contribute to obesity and related complications. The intracellular steps of chylomicron biogenesis and transport from the Endoplasmic Reticulum (ER) to the Golgi complex have been described, but the mechanisms for post-Golgi transport and secretion of chylomicrons have not been identified. Using a newly generated Dennd5b-/- mouse, we demonstrate an essential role for this gene in Golgi to plasma membrane transport of chylomicron secretory vesicles. In mice, loss of Dennd5b results in resistance to western diet induced obesity, changes in plasma lipids, and reduced aortic atherosclerosis. In humans, two independent exome sequencing studies reveal that a common DENND5B variant, p.(R52K), is correlated with body mass index. These studies establish an important role for DENND5B in post-Golgi chylomicron secretion and a subsequent influence on body composition and peripheral lipoprotein metabolism.


Body Mass Index , Death Domain Receptor Signaling Adaptor Proteins/physiology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Intestinal Absorption , Obesity/prevention & control , Triglycerides/metabolism , Animals , Biological Transport , Diet, High-Fat/adverse effects , Female , Lipid Metabolism , Male , Mice , Mice, Knockout , Obesity/etiology , Obesity/metabolism
3.
Dev Dyn ; 248(5): 351-362, 2019 05.
Article En | MEDLINE | ID: mdl-30884041

BACKGROUND: The DENND1A has been identified as a guanine nucleotide exchange factor for small GTPase Rab35, which functions in endocytic trafficking to mediate the recycling of selective cargos. Genetic alterations within the DENND1A gene have been implicated in human disease such as polycystic ovary syndrome (PCOS). However, the role of DENND1A in developmental and reproductive processes is largely unknown. RESULTS: Using Dennd1a gene knockout mice, we uncovered that homogeneous Dennd1a-/- mutants died around embryonic day (E) 14.5. The brain of Dennd1a-/- embryos exhibited defects, partially attributed to the dysregulation of cell division and survival in the telencephalon. The transcription of Fgf8 mRNA was ectopically elevated in the dorsal midline of telencephalon, concomitant with a decrease of active ß-catenin and Axin2 in the brain of Dennd1a-/- embryos. During liver morphogenesis, the ablation of Dennd1a impaired hepatic cell proliferation, the differentiation of hepatocyte, and hepatic hematopoiesis. In addition, loss of Dennd1a also affected the development of primordial germ cells. CONCLUSIONS: We demonstrate that Dennd1a, a susceptibility gene for PCOS, is essential for embryogenesis, probably through the mediation of endocytic recycling of selective cargos that are involved in cell signaling crucial for the development of multiple embryonic organ systems.


Death Domain Receptor Signaling Adaptor Proteins/physiology , Embryonic Development , Guanine Nucleotide Exchange Factors/physiology , Animals , Cell Division , Cell Survival , Death Domain Receptor Signaling Adaptor Proteins/genetics , Female , Genetic Predisposition to Disease , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Knockout/embryology , Polycystic Ovary Syndrome/genetics , Telencephalon/cytology
4.
BMB Rep ; 52(2): 119-126, 2019 Feb.
Article En | MEDLINE | ID: mdl-30638181

The tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) initiates the extrinsic apoptotic pathway through formation of the death-inducing signaling complex (DISC), followed by activation of effector caspases. TRAIL receptors are composed of death receptors (DR4 and DR5), decoy receptors (DcR1 and DcR2), and osteoprotegerin. Among them, only DRs activate apoptotic signaling by TRAIL. Since the levels of DR expressions are higher in cancer cells than in normal cells, TRAIL selectively activates apoptotic signaling pathway in cancer cells. However, multiple mechanisms, including down-regulation of DR expression and pro-apoptotic proteins, and up-regulation of anti-apoptotic proteins, make cancer cells TRAIL-resistant. Therefore, many researchers have investigated strategies to overcome TRAIL resistance. In this review, we focus on protein regulation in relation to extrinsic apoptotic signaling pathways via ubiquitination. The ubiquitin proteasome system (UPS) is an important process in control of protein degradation and stabilization, and regulates proliferation and apoptosis in cancer cells. The level of ubiquitination of proteins is determined by the balance of E3 ubiquitin ligases and deubiquitinases (DUBs), which determine protein stability. Regulation of the UPS may be an attractive target for enhancement of TRAIL-induced apoptosis. Our review provides insight to increasing sensitivity to TRAIL-mediated apoptosis through control of post-translational protein expression. [BMB Reports 2019; 52(2): 119-126].


Apoptosis/physiology , Deubiquitinating Enzymes/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Caspases/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/physiology , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/physiology , Tumor Necrosis Factor Decoy Receptors/physiology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Mol Biol Rep ; 45(6): 2193-2199, 2018 Dec.
Article En | MEDLINE | ID: mdl-30232779

Single nucleotide polymorphisms (SNPs), the most common genetic variations in human genome, can manage the predisposition of certain complex diseases or situations such as obesity. Genetic polymorphisms also play an important role as they can impact a population's susceptibility to being overweight or obese and developing related chronic complications such as hypertension, coronary heart disease, diabetes and cancer. The present study comprised of 193 unrelated healthy volunteers (120 females and 73 males) with Turkish origin. Only female adolescents (n = 110) were divided into 2 categories according to their BMI values as overweight (BMI ≥ 25) and normal (18.5 < BMI < 25) according to WHO classification. Genomic DNA was isolated from venous blood samples and genotyping of DENND1A rs10818854 and CYP19A1 rs2414096 variants was performed on Roche Light Cycler 2.0 Real-Time PCR platform. Serum hormone levels were analyzed by Electrochemiluminescent Immunoassay (ECLIA; Roche diagnostics). The genotype distributions were consistent with the Hardy-Weinberg equilibrium for both SNPs in the studied population (p > 0.05). The genotype distribution of DENND1A rs10818854 was determined for the first time in Turkish population and the variant allele frequency was found as 0.095. According to reduced sex hormone-binding globulin levels and increased free androgen index in the present study, obesity was linked with hyperandrogenism in female subjects. Both polymorphisms were investigated as potential genetic susceptibility markers for obesity and neither DENND1A nor CYP19A1 showed any associations.


Aromatase/genetics , Death Domain Receptor Signaling Adaptor Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Obesity/genetics , Adolescent , Adult , Alleles , Aromatase/metabolism , Body Mass Index , Case-Control Studies , Child , Death Domain Receptor Signaling Adaptor Proteins/physiology , Female , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genotype , Guanine Nucleotide Exchange Factors/physiology , Humans , Male , Overweight , Polymorphism, Single Nucleotide/genetics , Preliminary Data , Turkey , Young Adult
6.
J Cell Sci ; 130(22): 3779-3787, 2017 Nov 15.
Article En | MEDLINE | ID: mdl-29142064

The PIDDosome is often used as the alias for a multi-protein complex that includes the p53-induced death domain protein 1 (PIDD1), the bipartite linker protein CRADD (also known as RAIDD) and the pro-form of an endopeptidase belonging to the caspase family, i.e. caspase-2. Yet, PIDD1 variants can also interact with a number of other proteins that include RIPK1 (also known as RIP1) and IKBKG (also known as NEMO), PCNA and RFC5, as well as nucleolar components such as NPM1 or NCL. This promiscuity in protein binding is facilitated mainly by autoprocessing of the full-length protein into various fragments that contain different structural domains. As a result, multiple responses can be mediated by protein complexes that contain a PIDD1 domain. This suggests that PIDD1 acts as an integrator for multiple types of stress that need instant attention. Examples are various types of DNA lesion but also the presence of extra centrosomes that can foster aneuploidy and, ultimately, promote DNA damage. Here, we review the role of PIDD1 in response to DNA damage and also highlight novel functions of PIDD1, such as in centrosome surveillance and scheduled polyploidisation as part of a cellular differentiation program during organogenesis.


Centrosome/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Animals , Apoptosis , CRADD Signaling Adaptor Protein/physiology , Caspase 2/physiology , Cell Differentiation , DNA Damage , Humans , Multiprotein Complexes/physiology , Nucleophosmin , Polyploidy
7.
Tumour Biol ; 37(8): 10447-57, 2016 Aug.
Article En | MEDLINE | ID: mdl-26846109

p53-induced death domain protein (PIDD) facilitates p53-dependent apoptosis through the interaction with components of the death receptor signaling pathways. However, the role of PIDD in hepatocellular carcinoma (HCC) development remains unknown. In this study, we investigated the expression pattern of PIDD in clinical HCC samples and adjacent non-cancerous tissues using immunohistochemistrical and Western blot analyses. The results showed that PIDD was lowly expressed in HCC tissues and HCC cell lines, compared with the adjacent non-tumorous tissues and LO2 normal hepatocytes. In addition, clinicopathological analysis showed that the expression of PIDD was closely related with multiple clinicopathological variables, such as American Joint Committee on Cancer (AJCC) stage, AFP, and poor prognosis of HCC. Univariate and multivariate survival analyses demonstrated that PIDD could serve as an independent prognostic factor to predict the survival of HCC patients. We used serum starvation-refeeding experiment to explore the involvement of PIDD in HCC cell cycle regulation. We found that PIDD was accumulated in growth-arrested HCC cells and was progressively decreased when cells entered into S phase. Moreover, flow cytometry and cell counting kit-8 (CCK-8) assays indicated that depleting the expression of PIDD could facilitate cell cycle progression and accelerate cell proliferation in HepG2 cells, while overexpression of PIDD could result in cell cycle arrest at G1 phase and hinder the cell proliferation in Hep3B cells. Finally, flow cytometry revealed that overexpression of PIDD slightly increased the apoptosis of HCC cells. Taken together, we concluded that PIDD may be a valuable prognostic marker and promising therapeutic target of HCC.


Carcinoma, Hepatocellular/pathology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Liver Neoplasms/pathology , Neoplasm Proteins/physiology , Adult , Aged , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Division , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/analysis , Death Domain Receptor Signaling Adaptor Proteins/antagonists & inhibitors , Female , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/antagonists & inhibitors , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Young Adult
8.
Math Biosci ; 270(Pt A): 126-34, 2015 Dec.
Article En | MEDLINE | ID: mdl-26524142

This paper is a continuation of our previous work (Huang and Yong, 2013) for simplifying the Fas signaling-induced apoptotic pathway identified by Hua et al. (2005) for human tumor T cells. The previous paper studied the downstream intracelluar-signaling subsystem, while the present one is concerned with the upstream death-inducing signaling complex (DISC) subsystem. Under the assumption that the bind of Fas-associated death domains and FLICE-inhibitory proteins to the DISC is much faster than that of the initiator procaspases, we greatly simplify the upstream subsystem from 35 reactions with 26 species to 6 reactions with 9 species by adopting the classical and recently justified partial equilibrium approximation method. Numerical simulations show that the simplified model is in an excellent agreement with the original model. Most importantly, the simplified model clearly reveals the key reactants and dominated pathways in the Fas signaling process, and thus provides new insights into the apoptosis.


Apoptosis/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Humans , Kinetics , Mathematical Concepts , Models, Biological , Signal Transduction
9.
Braz. j. med. biol. res ; 48(4): 292-298, 4/2015.
Article En | LILACS | ID: lil-744365

Programmed necrosis or necroptosis is an alternative form of cell death that is executed through a caspase-independent pathway. Necroptosis has been implicated in many pathological conditions. Genetic or pharmacological inhibition of necroptotic signaling has been shown to confer neuroprotection after traumatic and ischemic brain injury. Therefore, the necroptotic pathway represents a potential target for neurological diseases that are managed by neurosurgeons. In this review, we summarize recent advances in the understanding of necroptotic signaling pathways and explore the role of necroptotic cell death in craniocerebral trauma, brain tumors, and cerebrovascular diseases.


Humans , Apoptosis/physiology , Brain Injuries/therapy , Cerebrovascular Disorders/therapy , Necrosis/therapy , Receptors, Death Domain/physiology , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Death , Cerebrovascular Disorders/pathology , Cerebrovascular Disorders/physiopathology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Hydroxycholesterols/pharmacology , Necrosis/physiopathology , Neuroprotective Agents/antagonists & inhibitors , Signal Transduction/physiology , Toll-Like Receptors/physiology
10.
Braz J Med Biol Res ; 48(4): 292-8, 2015 Apr.
Article En | MEDLINE | ID: mdl-25714887

Programmed necrosis or necroptosis is an alternative form of cell death that is executed through a caspase-independent pathway. Necroptosis has been implicated in many pathological conditions. Genetic or pharmacological inhibition of necroptotic signaling has been shown to confer neuroprotection after traumatic and ischemic brain injury. Therefore, the necroptotic pathway represents a potential target for neurological diseases that are managed by neurosurgeons. In this review, we summarize recent advances in the understanding of necroptotic signaling pathways and explore the role of necroptotic cell death in craniocerebral trauma, brain tumors, and cerebrovascular diseases.


Apoptosis/physiology , Brain Injuries/therapy , Cerebrovascular Disorders/therapy , Necrosis/therapy , Receptors, Death Domain/physiology , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Death , Cerebrovascular Disorders/pathology , Cerebrovascular Disorders/physiopathology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Humans , Hydroxycholesterols/pharmacology , Necrosis/physiopathology , Neuroprotective Agents/antagonists & inhibitors , Signal Transduction/physiology , Toll-Like Receptors/physiology
11.
Apoptosis ; 20(2): 235-9, 2015 Feb.
Article En | MEDLINE | ID: mdl-25451007

Death-inducing signaling complex (DISC) is a platform for the activation of initiator caspase in extrinsic apoptosis. Assembly of DISC is accomplished by two different types of homotypic interaction: one is between death domains (DDs) of a death receptor and FADD, and the other is between death effecter domains (DEDs) of FADD, procaspase-8/-10 and cFLIP. Recent biochemical investigations on the stoichiometry of DISC have revealed that single-DED-containing FADD exists in DISC in a substantially lower abundance than the sum of tandem-DEDs-containing components that are procaspase-8 and cFLIP. In addition, the homology models of the tandem DEDs in procaspase-8 and cFLIP show that two different interaction faces, H1-H4 face and H2-H5 face, are exposed for possible inter-molecular DED-DED interactions. These recent findings led to a proposal of the DED chain model for the interactions between FADD, procaspase-8 and cFLIP in DISC. This emerging view provides new insights on the topology of DED-DED network in DISC and furthermore on how procaspase-8 and cFLIP cluster for dimerization and proteolytic activation.


Apoptosis , Death Domain Receptor Signaling Adaptor Proteins/physiology , Animals , Humans , Inflammation/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , Signal Transduction
12.
Apoptosis ; 20(2): 124-35, 2015 Feb.
Article En | MEDLINE | ID: mdl-25398537

Apoptosis is an important process to maintain cellular homeostasis. Deregulated apoptosis has linked to a number of diseases, such as inflammatory diseases, neurodegenerative disorder, and cancers. A major signaling complex in the death receptor signaling pathway leading to apoptosis is death-induced signaling complex (DISC), which is regulated mainly by death effector domain (DED)-containing proteins. There are seven DED-containing proteins in human, including FADD, c-FLIP, caspase-8, caspase-10, DEDD, DEDD2, and PEA-15. The main players in DISC formation employ tandem DEDs for regulating signaling complex formation. The regulatory mechanism of signaling complex formation is important and yet remains unclear. Interestingly, three caspase recruitment domain (CARD)-containing members, which belong to the same DD superfamily as DED-containing proteins, also contains similar tandem CARDs. Recent structural studies have shown that tandem CARDs are essential for the formation of a helical signaling complex. This review summarizes recent structural studies on DED-containing proteins and especially discusses the studies on tandem DEDs and tandem CARDs, which suggest new mechanisms of signaling complex assembly.


Apoptosis , CARD Signaling Adaptor Proteins/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , CARD Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/physiology , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Signal Transduction , Structural Homology, Protein
13.
Apoptosis ; 20(2): 151-6, 2015 Feb.
Article En | MEDLINE | ID: mdl-25398536

Inflammasomes are oligomeric protein complexes assembled through interactions among the death domain superfamily members, in particular the CARD and PYD domains. Recent progress has shed lights on how the ASC PYD can polymerize to form filaments using multiple domain:domain interfaces, and how the caspase4 CARD can recognize LPS to activate the non-classical inflammasome pathway. Comprehensive understanding of the molecular mechanisms of inflammasome activation and assembly require more extensive structural and biophysical dissection of the inflammasome components and complexes, in particular additional CARD or PYD filaments. Because of the variations in death domain structures and complexes observed so far, future work will undoubtedly shed lights on the mechanisms of inflammasome assembly as well as more surprises on the versatile structure and function of the death domain superfamily.


Inflammasomes/physiology , Animals , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/physiology , Humans , Inflammasomes/chemistry , Models, Molecular , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Structural Homology, Protein
14.
Annu Rev Cell Dev Biol ; 30: 337-56, 2014.
Article En | MEDLINE | ID: mdl-25150011

Cell turnover is a fundamental feature in metazoans. Cells can die passively, as a consequence of severe damage to their structural integrity, or actively, owing to a more confined biological disruption such as DNA damage. Passive cell death is uncontrolled and often harmful to the organism. In contrast, active cell death is tightly regulated and serves to support the organism's life. Apoptosis-the primary form of regulated cell death-is relatively well defined. Necroptosis-an alternative, distinct kind of regulated cell death discovered more recently-is less well understood. Apoptosis and necroptosis can be triggered either from within the cell or by extracellular stimuli. Certain signaling components, including several death ligands and receptors, can regulate both processes. Whereas apoptosis is triggered and executed via intracellular proteases called caspases, necroptosis is suppressed by caspase activity. Here we highlight current understanding of the key signaling mechanisms that control regulated cell death.


Cell Death/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/physiology , Caspases/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Enzyme Activation , Humans , Models, Biological , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Death Domain/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factors/physiology
15.
Exerc Immunol Rev ; 20: 117-34, 2014.
Article En | MEDLINE | ID: mdl-24974724

Physical exercise is well known to affect leukocyte numbers and function. While regular exercise training has been shown to enhance specific immune functions, acute bouts of intensive exercise often lead to a pro-inflammatory response accompanied by a transient lymphocytopenia and neutrophilia. It can be assumed, that lymphocytopenia can be attributed at least partially to an enhanced lymphocyte apoptosis. In contrast, regulation of neutrophil apoptosis after exercise remains controversial since studies demonstrated both an up-regulation as well as a down-regulation of cell death. However, these discrepancies may be due to differences in exercise protocols, subjects' fitness levels, and to different methodological approaches. Two major signalling pathways of exercise induced apoptosis have been identified. First the external receptor mediated pathway using death receptors, and second the internal, oxidative-mediated pathway which encompasses the mitochondria. Potential apoptosis modulating mediators are reactive oxygen species (ROS), glucocorticoids and cytokines which are part of the systemic inflammatory response evoked after acute intensive exercise. Finally, the physiological impact and clinical relevance of leukocyte apoptosis will be discussed. On the one hand, exercise-induced apoptosis might be a mechanism to remove activated and potentially autoreactive immune cells. On the other hand, apoptosis might be a regulatory mechanism which is necessary for tissue reorganization and adaptational training processes.


Apoptosis , Exercise/physiology , Inflammation/etiology , Leukocytes/pathology , Lymphopenia/etiology , Cytokines/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Glucocorticoids/physiology , Humans , Inflammation/blood , Inflammation/pathology , Lymphocytes/pathology , Lymphopenia/pathology , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Stress, Physiological/immunology , fas Receptor/physiology
16.
Math Biosci ; 246(1): 27-37, 2013 Nov.
Article En | MEDLINE | ID: mdl-24075944

Apoptosis is one of the most basic biological processes. In apoptosis, tens of species are involved in many biochemical reactions with times scales of widely differing orders of magnitude. By the law of mass action, the process is mathematically described with a large and stiff system of ODEs (ordinary differential equations). The goal of this work is to simplify such systems of ODEs with the PEA (partial equilibrium approximation) method. In doing so, we propose a general framework of the PEA method together with some conditions, under which the PEA method can be justified rigorously. The main condition is the principle of detailed balance for fast reactions as a whole and the framework provides some meaningful physical insights of the full chemical kinetics. With the justified method as a tool, we simplify the Fas-signaling pathway model due to Hua et al. [6] under the empirical assumption that nine reactions therein can be well regarded as relatively fast. This paper reports our simplification, together with numerical results which confirm the reliability of both our simplified model and the empirical assumption.


Apoptosis/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Models, Theoretical , Signal Transduction/physiology , Humans
17.
J Mol Neurosci ; 48(1): 97-110, 2012 Sep.
Article En | MEDLINE | ID: mdl-22678883

The potential effects of alternative splicing of death-domain expressing genes and neuronal death have not been determined in Alzheimer's disease (AD). We analyzed DENN/MADD/IG20 (DMI), the complex of four splice variants. IG20 is known to be involved in cell death and the DENN/MADD splice variant (DM-SV) in cell survival in non-neural systems. DENN/MADD (DM) and DENN/MADD splice variant 2 were also included. Using SH-SY5Y human neuroblastoma cultures exposed to high concentrations of oligomeric Aß peptides (oAß) as a model for neuronal death, there was initially an increased ratio of DM-SV to IG20 (DM-SV/IG20) and knockdown of DMI SVs including DM-SV with antisense DNA then increased cell death. Cultures transfected with small interfering RNAs (siRNAs) specific to subsets of DMI SVs but sparing DM-SV increased the DM-SV/IG20 ratio resulting in a reduction of cell death in the presence of oAß. Effects on cell survival of DM and DM SV2, the other two SVs expressed in the CNS, are less clear. Compared to normal controls, alternative splicing changes in the CNS of AD patients during disease progression resulted in altered ratios of all of the SVs in a pattern over an extended time that mirrored that of the cultures, and coincided with the accumulation of endogenous, dimeric Aß (dAß). Thus, DM-SV may be required for neuronal survival by protecting against oAß neurotoxicity, and IG20 may contribute to selective neuronal vulnerability in AD.


Alternative Splicing/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cell Death/genetics , Death Domain Receptor Signaling Adaptor Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Aged, 80 and over , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Cell Death/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/genetics , Death Domain Receptor Signaling Adaptor Proteins/physiology , Dimerization , Female , Guanine Nucleotide Exchange Factors/physiology , Humans , Male , Neurites/pathology , Neurites/physiology , Neuroblastoma , Neurons/pathology , Neurons/physiology , RNA, Small Interfering/genetics
18.
Cancer Res ; 72(13): 3238-50, 2012 Jul 01.
Article En | MEDLINE | ID: mdl-22719072

Epithelial-to-mesenchymal transition (EMT), a crucial developmental program, contributes to cancer invasion and metastasis. In this study, we show that death-effector domain-containing DNA-binding protein (DEDD) attenuates EMT and acts as an endogenous suppressor of tumor growth and metastasis. We found that expression levels of DEDD were conversely correlated with poor prognosis in patients with breast and colon cancer. Both in vitro and in vivo, overexpression of DEDD attenuated the invasive phenotype of highly metastatic cells, whereas silencing of DEDD promoted the invasion of nonmetastatic cells. Via direct interaction with the class III PI-3-kinase (PI3KC3)/Beclin1, DEDD activated autophagy and induced the degradation of Snail and Twist, two master regulators of EMT. The DEDD-PI3KC3 interaction led to stabilization of PI3KC3, which further contributed to autophagy and the degradation of Snail and Twist. Together, our findings highlight a novel mechanism in which the intracellular signaling protein DEDD functions as an endogenous tumor suppressor. DEDD expression therefore may represent a prognostic marker and potential therapeutic target for the prevention and treatment of cancer metastasis.


Autophagy , Breast Neoplasms/pathology , DNA-Binding Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Epithelial-Mesenchymal Transition , Phosphatidylinositol 3-Kinases/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Division/physiology , Cell Line, Tumor , DNA-Binding Proteins/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Protein Binding
19.
Eur J Cancer ; 48(17): 3288-99, 2012 Nov.
Article En | MEDLINE | ID: mdl-22456178

Most gastric cancer cells are resistant to tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). Since TRAIL resistance is associated with lipid rafts, in which both death receptors and epidermal growth factor receptors (EGFR) are enriched, our aim is to identify how lipid raft-regulated receptor redistribution influences the sensitivity of TRAIL in gastric cancer cells. In TRAIL-resistant gastric cancer cells, TRAIL did not induce effective death-inducing signalling complex (DISC) formation in lipid rafts, accompanied with EGFR translocation into lipid rafts, and activation of EGFR pathway. Knockdown of casitas B-lineage lymphoma-b (Cbl-b) enhanced TRAIL-induced apoptosis by promoting DISC formation in lipid rafts. However, knockdown of Cbl-b also enhanced EGFR translocation into lipid rafts and EGFR pathway activation induced by TRAIL. Either using inhibitors of EGFR or depletion of EGFR with small interfering RNA (siRNA) prevented EGFR pathway activation, and thus increased TRAIL-induced apoptosis, especially in Cbl-b knockdown clones. Taken together, TRAIL-induced EGFR activation through Cbl-b-regulated EGFR redistribution in lipid rafts antagonised TRAIL-induced apoptosis. The contribution of DISC formation and the inhibition of EGFR signal triggered in lipid rafts are both essential for increasing the sensitivity of gastric cancer cells to TRAIL.


Adaptor Proteins, Signal Transducing/physiology , Apoptosis/drug effects , ErbB Receptors/metabolism , Membrane Microdomains/metabolism , Proto-Oncogene Proteins c-cbl/physiology , Stomach Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/physiology , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Humans , Protein Transport/drug effects , Stomach Neoplasms/pathology
20.
J Biol Chem ; 287(15): 12455-68, 2012 Apr 06.
Article En | MEDLINE | ID: mdl-22362782

Autophagy and apoptosis are two evolutionarily conserved processes that regulate cell fate in response to cytotoxic stress. However, the functional relationship between these two processes remains far from clear. Here, we demonstrate an autophagy-dependent mechanism of caspase-8 activation and initiation of the apoptotic cascade in response to SKI-I, a pan-sphingosine kinase inhibitor, and bortezomib, a proteasome inhibitor. Autophagy is induced concomitantly with caspase-8 activation, which is responsible for initiation of the caspase cascade and the mitochondrial amplification loop that is required for full execution of apoptosis. Inhibition of autophagosome formation by depletion of Atg5 or Atg3 results in a marked suppression of caspase-8 activation and apoptosis. Although caspase-8 self-association depends on p62/SQSTM1, its self-processing requires the autophagosomal membrane. Caspase-8 forms a complex with Atg5 and colocalizes with LC3 and p62. Moreover, FADD, an adaptor protein for caspase-8 activation, associates with Atg5 on Atg16L- and LC3-positive autophagosomal membranes and loss of FADD suppresses cell death. Taken together, these results indicate that the autophagosomal membrane serves as a platform for an intracellular death-inducing signaling complex (iDISC) that recruits self-associated caspase-8 to initiate the caspase-8/-3 cascade.


Apoptosis , Autophagy , Caspase 8/metabolism , Cell Membrane/metabolism , Death Domain Receptor Signaling Adaptor Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Caspase 3/metabolism , Cell Membrane/enzymology , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation , Fas-Associated Death Domain Protein/metabolism , Gene Knockout Techniques , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hydrazines/pharmacology , Leukemia, Myeloid, Acute , Lysosomal Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Multimerization , Protein Transport , Pyrazoles/pharmacology , Sequestosome-1 Protein , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
...