Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
ACS Omega ; 6(31): 20139-20148, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34373846

ABSTRACT

Severe acute respiratory coronavirus-2 (SARS-CoV-2) is a novel viral pathogen and therefore a challenge to accurately diagnose infection. Asymptomatic cases are common and so it is difficult to accurately identify infected cases to support surveillance and case detection. Diagnostic test developers are working to meet the global demand for accurate and rapid diagnostic tests to support disease management. However, the focus of many of these has been on molecular diagnostic tests, and more recently serologic tests, for use in primarily high-income countries. Low- and middle-income countries typically have very limited access to molecular diagnostic testing due to fewer resources. Serologic testing is an inappropriate surrogate as the early stages of infection are not detected and misdiagnosis will promote continued transmission. Detection of infection via direct antigen testing may allow for earlier diagnosis provided such a method is sensitive. Leading SARS-CoV-2 biomarkers include spike protein, nucleocapsid protein, envelope protein, and membrane protein. This research focuses on antibodies to SARS-CoV-2 spike protein due to the number of monoclonal antibodies that have been developed for therapeutic research but also have potential diagnostic value. In this study, we assessed the performance of antibodies to the spike glycoprotein, acquired from both commercial and private groups in multiplexed liquid immunoassays, with concurrent testing via a half-strip lateral flow assays (LFA) to indicate antibodies with potential in LFA development. These processes allow for the selection of pairs of high-affinity antispike antibodies that are suitable for liquid immunoassays and LFA, some of which with sensitivity into the low picogram range with the liquid immunoassay formats with no cross-reactivity to other coronavirus S antigens. Discrepancies in optimal ranking were observed with the top pairs used in the liquid and LFA formats. These findings can support the development of SARS-CoV-2 LFAs and diagnostic tools.

2.
Immunol Cell Biol ; 95(4): 340-346, 2017 04.
Article in English | MEDLINE | ID: mdl-27974746

ABSTRACT

The use of antibody therapy for cancer has steadily increased in recent years and has become standard treatment for numerous tumor types. It is now appreciated that the clinical activity of these antibodies relies upon their specific interactions with Fc receptors in addition to the well-studied target-binding region. The interactions mediated by antibody Fc domains can strongly affect the functional outcome of antibody therapy. The Fc portion of an antibody defines its interaction with numerous immune cells and has become an intense area of research as selecting the optimal Fc can greatly enhance the activity as well as mechanism of action of therapeutic antibodies. Recent advances in antibody engineering have enabled the development of antibodies that have altered Fc receptor interactions to take advantage of these findings. Engineering the Fc can fulfill diverse functions such as enhancing effector function for killing of tumor cells or depletion of unwanted immune subsets, enhancing agonist receptor signaling on particular immune cells or eliminating interaction with Fc receptors to avoid cellular depletion or toxicity in normal tissues. This review highlights important data and studies examining the role of Fc-Fc receptor interactions in therapeutic antibodies with a considerations for the future of engineered antibody therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Animals , Humans , Neoplasms/immunology , Protein Binding , Protein Engineering , Signal Transduction
3.
Cancer Cell ; 29(6): 820-831, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27265505

ABSTRACT

While engagement of the inhibitory Fcγ-receptor (FcγR) IIB is an absolute requirement for in vivo antitumor activity of agonistic mouse anti-CD40 monoclonal antibodies (mAbs), a similar requirement for human mAbs has been disputed. By using a mouse model humanized for its FcγRs and CD40, we revealed that FcγRIIB engagement is essential for the activity of human CD40 mAbs, while engagement of the activating FcγRIIA inhibits this activity. By engineering Fc variants with selective enhanced binding to FcγRIIB, but not to FcγRIIA, significantly improved antitumor immunity was observed. These findings highlight the necessity of optimizing the Fc domain for this class of therapeutic antibodies by using appropriate preclinical models that accurately reflect the unique affinities and cellular expression of human FcγR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , CD40 Antigens/agonists , Neoplasms/drug therapy , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/immunology , Cell Line, Tumor , Humans , Immunotherapy , Mice , Neoplasms/immunology , Xenograft Model Antitumor Assays
4.
Cancer Biol Ther ; 7(9): 1441-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18708753

ABSTRACT

Elevated activity of the eIF4F complex, which controls initiation of cap-dependent mRNA translation, has been linked to cancer progression. eIF4E recruitment to eIF4F is the rate limiting step of complex assembly and is regulated by eIF4E-Binding Proteins (4E-BPs). When stimulated, the mammalian Target of Rapamycin complex 1 (mTORC1) phosphorylates 4E-BP1, which then releases eIF4E. Hypoxia inhibits mTORC1 activity and therefore cap-dependent protein synthesis. To establish a novel genetic test of the role of eIF4F activity in regulating cell division and viability within hypoxic tumor microenvironments, we generated shRNA mediated 4E-BP1 knock-down in Rh30 rhabdomyosarcoma cells. 4E-BP1 knock-down relieved hypoxia-mediated inhibition of cycle progression in vitro and was correlated with increased expression of cyclin D1 and c-Myc. Xenograft tumors derived from these cells also displayed enhanced expression of cyclin D1 and c-Myc along with antiapoptotic genes encoding Bcl-x(L), and XIAP, and failed to develop the extensive necrotic zones and edema observed in control tumors. Surprisingly, 4E-BP1 knock-down also leads to a dramatic increase in aberrant mitoses in vivo and enhanced expression of Mad2 and securin. Thus, reduced expression of the negative regulator of eIF4E has significant effects on tumor development, and is associated with enhanced cell proliferation and survival.


Subject(s)
Cell Cycle , Cell Proliferation , Eukaryotic Initiation Factor-4E/metabolism , Hypoxia/metabolism , Neoplasms/metabolism , Animals , Cell Survival , Cellular Structures/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Immunohistochemistry , Mice , Mice, Nude , Neoplasms/genetics , RNA, Small Interfering/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
5.
Annu Rev Physiol ; 70: 51-71, 2008.
Article in English | MEDLINE | ID: mdl-17850210

ABSTRACT

Low oxygen (O2) levels are a naturally occurring feature of embryonic development, adult physiology, and diseases such as those of the cardiovascular system. Although many responses to O2 deprivation are mediated by hypoxia-inducible factors (HIFs), researchers are finding a growing number of HIF-independent pathways that promote O2 conformance and hypoxia tolerance. Here, we describe HIF-independent responses and how they impact cardiovascular tissue homeostasis.


Subject(s)
Cardiovascular System/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Signal Transduction/physiology , Animals , Homeostasis/physiology , Humans , Ischemia/metabolism
6.
J Clin Invest ; 117(9): 2385-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17786234

ABSTRACT

Increased cap-dependent mRNA translation rates are frequently observed in human cancers. Mechanistically, many human tumors often overexpress the cap binding protein eukaryotic translation initiation factor 4E (eIF4E), leading to enhanced translation of numerous tumor-promoting genes. In this issue of the JCI, Graff and colleagues describe potent antitumor effects using second-generation antisense oligonucleotides for eIF4E (see the related article beginning on page 2638). If their results are recapitulated in a clinical setting, this strategy will provide a promising antitumor therapy with broad-reaching applications.


Subject(s)
Neoplasms/genetics , Protein Biosynthesis/genetics , Animals , Down-Regulation , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy
7.
Cancer Metastasis Rev ; 26(2): 261-71, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17647111

ABSTRACT

Recent studies have described a small population of self-renewing and multipotent cells within tumors termed "cancer stem cells." These cells share many traits with somatic and embryonic stem cells and are thought to be responsible for driving tumor progression in a growing list of neoplastic diseases. Cells within solid tumors encounter hypoxia due to poor vascular function. Both long-standing and emerging data describe hypoxic effects on somatic and embryonic stem cells, and it is likely that hypoxia also has profound effects on cancer stem cells. These effects include the activation of pathways that induce the dedifferentiation of cancer cells, the maintenance of stem cell identity, and increased metastatic potential. Hypoxia may contribute to tumor progression by specifically impacting these pathways in cancer stem cells.


Subject(s)
Cell Hypoxia/physiology , Neoplasm Metastasis/physiopathology , Stem Cells/physiology , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasm Metastasis/pathology , Stem Cells/pathology
8.
Biochim Biophys Acta ; 1755(1): 25-36, 2005 May 25.
Article in English | MEDLINE | ID: mdl-15907590

ABSTRACT

CD95 (APO-1/Fas) is an important inducer of the extrinsic apoptosis signaling pathway and therapy induced apoptosis of many tumor cells has been linked to the activity of CD95. Changes in the expression of CD95 and/or its ligand CD95L are frequently found in human cancer. The downregulation or mutation of CD95 has been proposed as a mechanism by which cancer cells avoid destruction by the immune system through reduced apoptosis sensitivity. CD95 has therefore been viewed as a tumor suppressor. Furthermore, increased CD95L concentration in tumor patients has been linked to tumor cells killing infiltrating lymphocytes in a process called "the tumor counter-attack". Recent data have illuminated unknown activities of CD95 in tumor cells with downregulated or mutated CD95 in the presence of increased CD95L. Under these conditions the stimulation of CD95 signals nonapoptotic pathways, activating NF-kappaB and MAP kinases for example, which may result in the induction of tumorigenic or prosurvival genes. A new model of CD95 functions is proposed in which CD95 is converted from a tumor suppressor to a tumor promotor by a single point mutation in one of the CD95 alleles, a situation frequently found in advanced human cancer, resulting in apoptosis resistance and activation of tumorigenic pathways.


Subject(s)
Neoplasms/metabolism , fas Receptor/physiology , Animals , Apoptosis , Disease Progression , Down-Regulation , Fas Ligand Protein , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred MRL lpr , Models, Biological , Mutation , Neoplasms/therapy , Signal Transduction
9.
EMBO Rep ; 5(11): 1084-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15514680

ABSTRACT

Mutations in the death domain of the death receptor CD95 (APO-1/Fas) cause lymphoproliferation and autoimmune disease in both lpr(cg) mice and in patients with autoimmune lymphoproliferative syndrome (ALPS) type Ia. By testing lymphocytes from ALPS type Ia patients, comparing heterozygous with homozygous lpr(cg) mice and coexpressing wild-type and mutant CD95 receptors, we demonstrate that induction of apoptosis requires two wild-type alleles of CD95. By contrast, nuclear factor-kappaB (NF-kappaB) can be fully activated in cells expressing both a mutant and a wild-type CD95 allele, suggesting different thresholds to activate the two signalling pathways. This was confirmed by testing lymphocytes from heterozygous lpr mice, which showed reduced sensitivity to CD95-mediated apoptosis but normal activation of NF-kappaB when compared with wild-type mice. Mutations in CD95 may eliminate the tumour-suppressive function of CD95, at the same time allowing induction of survival or proliferative pathways, which could contribute to the increased risk for lymphoma seen in ALPS type Ia patients.


Subject(s)
Apoptosis , NF-kappa B/metabolism , Signal Transduction , fas Receptor/biosynthesis , Alleles , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chloramphenicol O-Acetyltransferase/metabolism , Enzyme-Linked Immunosorbent Assay , Exons , Heterozygote , Homozygote , Humans , Ligands , Lymphocytes/metabolism , Mice , Models, Biological , Mutation , Plasmids/metabolism , Spleen/cytology , Spleen/metabolism , Time Factors
10.
Cell Cycle ; 3(10): 1235-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15467462

ABSTRACT

Most members of the death receptor family including CD95 (APO-1/Fas) have been shown to induce both apoptosis as well as non-apoptotic pathways depending on the tissue and the circumstances. One of the non-apoptotic pathways emanating from CD95, activation of NF-kappaB, has recently been demonstrated to regulate invasiveness of apoptosis resistant tumor cells. In contrast, activation of NF-kappaB in apoptosing cells is believed to be suppressed due to cleavage of various NF-kappaB pathway components by active caspases that execute apoptosis. We now present data demonstrating that in certain highly CD95 apoptosis sensitive cells NF-kappaB is robustly activated. In fact overexpression of apoptosis inhibitors such as Bcl-2 or c-FLIPL in these cells results in decreased activation of NF-kappaB through CD95. We propose a model in which NF-kappaB is generally activated in certain cells but may have different functions depending on whether cells are programmed to die or to survive.


Subject(s)
NF-kappa B/metabolism , Neoplasms/metabolism , Signal Transduction , fas Receptor/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , Down-Regulation/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Neoplasms/pathology
11.
J Biol Chem ; 279(45): 46742-7, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15345718

ABSTRACT

The death receptor CD95 (APO-1/Fas) induces apoptosis in many tissues. However, in apoptosis-resistant tumor cells, stimulation of CD95 induces up-regulation of a defined number of mostly anti-apoptotic genes, resulting in increased motility and invasiveness of tumor cells. The majority of these genes are known NF-kappaB target genes. We have identified one of the CD95-regulated genes as the serine/threonine kinase (SNF1/AMP kinase-related kinase (SNARK)), which is induced in response to various forms of metabolic stress. We demonstrate that up-regulation of SNARK in response to CD95 ligand and tumor necrosis factor alpha depends on activation of NF-kappaB. Overexpression of SNARK rendered tumor cells more resistant, whereas a kinase-inactive mutant of SNARK sensitized cells to CD95-mediated apoptosis. Furthermore, small interfering RNA-mediated knockdown of SNARK increased the sensitivity of tumor cells to CD95 ligand- and TRAIL-induced apoptosis. Importantly, cells with reduced expression of SNARK also showed reduced motility and invasiveness in response to CD95 engagement. SNARK therefore represents an NF-kappaB-regulated anti-apoptotic gene that contributes to the tumor-promoting activity of CD95 in apoptosis-resistant tumor cells.


Subject(s)
Apoptosis , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/physiology , fas Receptor/biosynthesis , Blotting, Western , Cell Death , Cell Line , Cell Line, Tumor , Cell Movement , Cell Survival , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , HeLa Cells , Humans , Mutagenesis, Site-Directed , Neoplasm Invasiveness , Phosphorylation , Plasmids/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
12.
EMBO J ; 23(15): 3175-85, 2004 Aug 04.
Article in English | MEDLINE | ID: mdl-15272306

ABSTRACT

The apoptosis-inducing death receptor CD95 (APO-1/Fas) controls the homeostasis of many tissues. Despite its apoptotic potential, most human tumors are refractory to the cytotoxic effects of CD95 ligand. We now show that CD95 stimulation of multiple apoptosis-resistant tumor cells by CD95 ligand induces increased motility and invasiveness, a response much less efficiently triggered by TNFalpha or TRAIL. Three signaling pathways resulting in activation of NF-kappaB, Erk1/2 and caspase-8 were found to be important to this novel activity of CD95. Gene chip analyses of a CD95-stimulated tumor cell line identified a number of potential survival genes and genes that are known to regulate increased motility and invasiveness of tumor cells to be induced. Among these genes, urokinase plasminogen activator was found to be required for the CD95 ligand-induced motility and invasiveness. Our data suggest that CD95L, which is found elevated in many human cancer patients, has tumorigenic activities on human cancer cells. This could become highly relevant during chemotherapy, which can cause upregulation of CD95 ligand by both tumor and nontumor cells.


Subject(s)
Apoptosis , Cell Movement , Membrane Glycoproteins/metabolism , Neoplasm Invasiveness/pathology , Neoplasms/metabolism , Neoplasms/pathology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Caspase 8 , Caspases/metabolism , Cell Movement/drug effects , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Fas Ligand Protein , Humans , MAP Kinase Signaling System , Membrane Glycoproteins/pharmacology , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , Urokinase-Type Plasminogen Activator/metabolism , fas Receptor/metabolism
13.
Oncogene ; 22(53): 8634-44, 2003 Nov 24.
Article in English | MEDLINE | ID: mdl-14634625

ABSTRACT

Apoptosis signaling is regulated and executed by specialized proteins that often carry protein/protein interaction domains. One of these domains is the death effector domain (DED) that is predominantly found in components of the death-inducing signaling complex, which forms at the members of the death receptor family following their ligation. Both proapoptotic- and antiapoptotic-DED-containing proteins have been identified, which makes these proteins exquisitely suited to the regulation of apoptosis. Aside from their pivotal role in the control of the apoptotic program, DED-containing proteins have recently been demonstrated to exert their influence on other cellular processes as well, including cell proliferation. These data highlight the multiple roles for the members of this family, suggesting that they are suited to control both life and death decisions of cells. Additionally, because they can act proapoptotically, antiapoptotically, or in the regulation of the cell cycle, this family of proteins may be excellent candidates for cancer therapy targets. Oncogene (2003) 22, 8634-8644. doi:10.1038/sj.onc.1207103


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , DNA-Binding Proteins , Intracellular Signaling Peptides and Proteins , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/physiology , Caspase 10 , Caspase 8 , Caspase 9 , Caspases/physiology , Death Domain Receptor Signaling Adaptor Proteins , Fas-Associated Death Domain Protein , Humans , Molecular Sequence Data , Neoplasms/therapy , Nuclear Proteins/physiology , Phosphoproteins/physiology , Proteins/physiology
14.
Semin Immunol ; 15(3): 185-93, 2003 Jun.
Article in English | MEDLINE | ID: mdl-14563117

ABSTRACT

CD95 (APO-1/Fas) has become the prototype of a death domain containing receptor and is the best studied member of the death receptors that activate the extrinsic apoptosis pathway. This pathway is initiated by recruitment and activation of caspase-8, an initiator caspase, in the death-inducing signaling complex (DISC) followed by direct cleavage of downstream effector caspases. In contrast, the intrinsic apoptosis pathway starts from within the cell either by direct activation of caspases or through intracellular changes such as DNA damage resulting in the release of a number of pro-apoptotic factors from the intermembrane space of mitochondria. The release of these factors results in the activation of another initiator caspase, caspase-9, and ultimately in the activation of effector caspases in a protein complex called the apoptosome. In recent years, it has become apparent that there is cross talk between the extrinsic and intrinsic pathway. In the death receptor pathway of apoptosis induction, the best characterized connection between the two pathways is the Bcl-2 family member Bid which translocates to mitochondria after cleavage by caspase-8 causing pro-apoptotic changes. Cells that die through CD95 without help from mitochondria are called Type I cells, whereas cells in which CD95-mediated death relies mostly on the intrinsic pathway are called Type II. This review focuses on recent developments in the delineation of the biochemistry and the physiological function of the two CD95 pathways.


Subject(s)
Mitochondria/metabolism , Signal Transduction , fas Receptor/metabolism , Animals , Fas Ligand Protein , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Models, Immunological , fas Receptor/immunology
15.
Proc Natl Acad Sci U S A ; 100(20): 11445-50, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14504390

ABSTRACT

CD95 type I and II cells differ in their dependence on mitochondria to execute apoptosis, because antiapoptotic members of the Bcl-2 family render only type II cells resistant to death receptor-induced apoptosis. They can also be distinguished by a more efficient formation of the death-inducing signaling complex in type I cells. We have identified a soluble form of CD95 ligand (S2) that is cytotoxic to type II cells but does not kill type I cells. By testing 58 tumor cell lines of the National Cancer Institute's anticancer drug-screening panel for apoptosis sensitivity to S2 and performing death-inducing signaling complex analyses, we determined that half of the CD95-sensitive cells are type I and half are type II. Most of the type I cell lines fall into a distinct class of tumor cells expressing mesenchymal-like genes, whereas the type II cell lines preferentially express epithelium-like markers. This suggests that type I and II tumor cells represent different stages of carcinogenesis that resemble the epithelial-mesenchymal transition. We then screened the National Cancer Institute database of >42,000 compounds for reagents with patterns of growth inhibition that correlated with either type I or type II cell lines and found that actin-binding compounds selectively inhibited growth of type I cells, whereas tubulin-interacting compounds inhibited growth of type II cells. Our analysis reveals fundamental differences in programs of gene expression between type I and type II cells and could impact the way actin- and microtubule-disrupting antitumor agents are used in tumor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/pathology , fas Receptor/immunology , Apoptosis , Cell Line , Drug Screening Assays, Antitumor , Humans , Neoplasms/immunology
16.
Cell ; 114(2): 148-50, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12887914

ABSTRACT

The tumor necrosis factor receptor 1 (TNFR1), a prototypic member of the death receptor family signals both cell survival and apoptosis. In this issue of Cell, report that apoptotic TNFR1 signaling proceeds via the sequential formation of two distinct complexes. Since the first complex can activate survival signals and influence the activity of the second complex, this mechanism provides a checkpoint to control the execution of apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Intracellular Signaling Peptides and Proteins , Proteins/metabolism , RNA-Binding Proteins , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Survival , Fas-Associated Death Domain Protein , Humans , Models, Biological , NF-kappa B/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Processing, Post-Translational , Proteins/genetics , Proteins/immunology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , TNF Receptor-Associated Factor 1 , Transcriptional Activation , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/metabolism
17.
Curr Opin Cell Biol ; 14(6): 721-6, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12473345

ABSTRACT

Caspases are well known for their role in the execution of the apoptotic program by cleaving specific target proteins, leading to the dismantling of the cell, as well as for mediating cytokine maturation. Recent work has highlighted novel non-apoptotic activities of apoptotic caspases. These reports indicate that caspases are much more versatile enzymes than we originally expected. In addition to regulating cell survival and cytokine maturation, caspases may be involved in regulating cell differentiation, cell proliferation, spreading and receptor internalization.


Subject(s)
Caspases/physiology , Animals , Apoptosis , Cell Cycle , Cell Division , Cell Movement , Cell Survival , Models, Biological , Receptors, Cell Surface/metabolism
19.
EMBO J ; 21(14): 3704-14, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12110583

ABSTRACT

Activation of the caspase cascade is a pivotal step in apoptosis and can occur via death adaptor-mediated homo-oligomerization of initiator procaspases. Here we show that c-FLIP(L), a protease-deficient caspase homolog widely regarded as an apoptosis inhibitor, is enriched in the CD95 death-inducing signaling complex (DISC) and potently promotes procaspase-8 activation through hetero-dimerization. c-FLIP(L) exerts its effect through its protease-like domain, which associates efficiently with the procaspase-8 protease domain and induces the enzymatic activity of the zymogen. Ectopic expression of c-FLIP(L) at physiologically relevant levels enhances procaspase-8 processing in the CD95 DISC and promotes apoptosis, while a decrease of c-FLIP(L) expression results in inhibition of apoptosis. c-FLIP(L) acts as an apoptosis inhibitor only at high ectopic expression levels. Thus, c-FLIP(L) defines a novel type of caspase regulator, distinct from the death adaptors, that can either promote or inhibit apoptosis.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Caspases/metabolism , Intracellular Signaling Peptides and Proteins , fas Receptor/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspase 9 , Cell Line , Dimerization , Enzyme Activation , Enzyme Precursors/metabolism , Humans , Precipitin Tests
20.
J Biol Chem ; 277(6): 4351-60, 2002 Feb 08.
Article in English | MEDLINE | ID: mdl-11733517

ABSTRACT

Apoptosis induction through CD95 (APO-1/Fas) critically depends on generation of active caspase-8 at the death-inducing signaling complex (DISC). Depending on the cell type, active caspase-8 either directly activates caspase-3 (type I cells) or relies on mitochondrial signal amplification (type II cells). In MCF7-Fas cells that are deficient for pro-caspase-3, even high amounts of caspase-8 produced at the DISC cannot directly activate downstream effector caspases without mitochondrial help. Overexpression of Bcl-x(L) in these cells renders them resistant to CD95-mediated apoptosis. However, activation of caspase-8 in control (vector) and Bcl-x(L) transfectants of MCF7-Fas cells proceeds with similar kinetics, resulting in a complete processing of cellular caspase-8. Most of the cytosolic caspase-8 substrates are not cleaved in the Bcl-x(L) protected cells, raising the question of how Bcl-x(L)-expressing MCF7-Fas cells survive large amounts of potentially cytotoxic caspase-8. We now demonstrate that active caspase-8 is initially generated at the DISC of both MCF7-Fas-Vec and MCF7-Fas-Bcl-x(L) cells and that the early steps of CD95 signaling such as caspase-8-dependent cleavage of DISC bound c-FLIP(L), caspase-8-dependent clustering, and internalization of CD95, as well as processing of pro-caspase-8 bound to mitochondria are very similar in both transfectants. However, events downstream of mitochondria, such as release of cytochrome c, only occur in the vector-transfected MCF7-Fas cells, and no in vivo caspase-8 activity can be detected in the Bcl-x(L)-expressing cells. Our data suggest that, in Bcl-x(L)-expressing MCF7-Fas cells, active caspase-8 is sequestered on the outer mitochondrial surface presumably by association with the protein "bifunctional apoptosis regulator" in a way that does not allow substrates to be cleaved, identifying a novel mechanism of regulation of apoptosis sensitivity by mitochondrial Bcl-x(L).


Subject(s)
Caspase Inhibitors , Mitochondria/enzymology , Proto-Oncogene Proteins c-bcl-2/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 8 , Caspase 9 , Humans , Precipitin Tests , Tumor Cells, Cultured , bcl-X Protein , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...