Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Omega ; 6(31): 20139-20148, 2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34373846

RESUMEN

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.
Oncoimmunology ; 7(8): e1466769, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30221069

RESUMEN

The use of inhibitory checkpoint blockade in the management of glioblastoma has been studied in both preclinical and clinical settings. TIGIT is a novel checkpoint inhibitor recently discovered to play a role in cancer immunity. In this study, we sought to determine the effect of anti-PD-1 and anti-TIGIT combination therapy on survival in a murine glioblastoma (GBM) model, and to elucidate the underlying immune mechanisms. Using mice with intracranial GL261-luc+ tumors, we found that TIGIT expression was upregulated on CD8+ and regulatory T cells (Tregs) in the brain compared to draining cervical lymph nodes (CLN) and spleen. We then demonstrated that treatment using anti-PD-1 and anti-TIGIT dual therapy significantly improved survival compared to control and monotherapy groups. The therapeutic effect was correlated with both increased effector T cell function and downregulation of suppressive Tregs and tumor-infiltrating dendritic cells (TIDCs). Clinically, TIGIT expression on tumor-infiltrating lymphocytes was shown to be elevated in patient GBM samples, suggesting that the TIGIT pathway may be a valuable therapeutic target. Expression of the TIGIT ligand, PVR, further portended a poor survival outcome in patients with low-grade glioma. We conclude that anti-TIGIT is an effective treatment strategy against murine GBM when used in combination with anti-PD-1, improving overall survival via modifications of both the T cell and myeloid compartments. Given evidence of PVR expression on human GBM cells, TIGIT presents as a promising immune therapeutic target in the management of these patients.

3.
SLAS Discov ; 23(7): 742-750, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29873570

RESUMEN

Enhancing antitumor activities of the human immune system is a clinically proven approach with the advent of monoclonal antibodies recognizing programmed cell death protein-1 (PD1) receptors on immune cell surfaces. Historically, using flow cytometry as a means to assess next-generation agent activities was underused, largely due to limits on cell number and assay sensitivity. Here, we leveraged an IntelliCyt high-throughput flow cytometry platform to monitor human dendritic cell maturation and lymphocyte proliferation in mixed lymphocyte reactions. Specifically, we established flow cytometry-based immunophenotyping and screening methodologies capable of measuring T-cell activation as a result of cell-associated antigens presented on dendritic cell surfaces, as indicated by cell proliferation, cytokine secretion, and surface marker expression. Together, the overall novelty of this 384-well platform is its capability to measure multiple functional readouts in one well and consistently evaluate large numbers of compounds in a single study, as well as its ability to show increased assay sensitivity requiring considerably fewer primary cells and less reagents compared to more traditional 96-well flow cytometry methods.


Asunto(s)
Células Dendríticas/metabolismo , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Activación de Linfocitos , Prueba de Cultivo Mixto de Linfocitos , Linfocitos T/metabolismo , Citocinas/metabolismo , Células Dendríticas/inmunología , Citometría de Flujo/métodos , Humanos , Activación de Linfocitos/inmunología , Linfocitos T/inmunología
4.
Immunol Cell Biol ; 95(4): 340-346, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27974746

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Animales , Humanos , Neoplasias/inmunología , Unión Proteica , Ingeniería de Proteínas , Transducción de Señal
5.
Cancer Cell ; 29(6): 820-831, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27265505

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD40/agonistas , Neoplasias/tratamiento farmacológico , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Línea Celular Tumoral , Humanos , Inmunoterapia , Ratones , Neoplasias/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Exp Cell Res ; 338(2): 251-60, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26302264

RESUMEN

Cellular levels of inhibitor of apoptosis (IAP) proteins are elevated in multiple human cancers and their activities often play a part in promoting cancer cell survival by blocking apoptotic pathways, controlling signal transduction pathways and contributing to resistance. These proteins function through interactions of their BIR (baculoviral IAP repeat) protein domains with pathway components and these interactions are endogenously antagonized by Smac/Diablo (second mitochondrial activator of caspases/direct IAP binding protein with low isoelectric point). This report describes development of synthetic smac mimetics (SM) and compares their binding, antiproliferative and anti-tumor activities. All dimeric antagonists inhibit in vitro smac tetrapeptide binding to recombinant IAP proteins, rescue IAP-bound caspase-3 activity and show anti-proliferative activity against human A875 melanoma cells. One heterodimeric SM, SM3, binds tightly to IAP proteins in vitro and slowly dissociates (greater than two hours) from these protein complexes compared to the other antagonists. In addition, in vitro SM anti-proliferation potency is influenced by ABCB1 transporter (ATP-binding cassette, sub-family B; MDR1, P-gp) activities and one antagonist, SM5, does not appear to be an ABCB1 efflux pump substrate. All dimeric smac mimetics inhibit the growth of human melanoma A875 tumors implanted in athymic mice at well-tolerated doses. One antagonist, SM4, shows broad spectrum in vivo anti-tumor activity and modulates known pharmacodynamic markers of IAP antagonism. These data taken together demonstrate the range of diverse dimeric IAP antagonist activities and supports their potential as anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Transporte Biológico/efectos de los fármacos , Caspasa 3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Mitocondriales/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Biomimética/métodos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Células HCT116 , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos
7.
Cancer Biol Ther ; 7(9): 1441-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18708753

RESUMEN

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.


Asunto(s)
Ciclo Celular , Proliferación Celular , Factor 4E Eucariótico de Iniciación/metabolismo , Hipoxia/metabolismo , Neoplasias/metabolismo , Animales , Supervivencia Celular , Estructuras Celulares/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoxia/genética , Inmunohistoquímica , Ratones , Ratones Desnudos , Neoplasias/genética , ARN Interferente Pequeño/metabolismo , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Annu Rev Physiol ; 70: 51-71, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17850210

RESUMEN

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.


Asunto(s)
Sistema Cardiovascular/metabolismo , Hipoxia/metabolismo , Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Homeostasis/fisiología , Humanos , Isquemia/metabolismo
9.
J Clin Invest ; 117(9): 2385-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17786234

RESUMEN

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.


Asunto(s)
Neoplasias/genética , Biosíntesis de Proteínas/genética , Animales , Regulación hacia Abajo , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia
10.
Cancer Metastasis Rev ; 26(2): 261-71, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17647111

RESUMEN

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.


Asunto(s)
Hipoxia de la Célula/fisiología , Metástasis de la Neoplasia/fisiopatología , Células Madre/fisiología , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metástasis de la Neoplasia/patología , Células Madre/patología
11.
Biochim Biophys Acta ; 1755(1): 25-36, 2005 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-15907590

RESUMEN

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.


Asunto(s)
Neoplasias/metabolismo , Receptor fas/fisiología , Animales , Apoptosis , Progresión de la Enfermedad , Regulación hacia Abajo , Proteína Ligando Fas , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Modelos Biológicos , Mutación , Neoplasias/terapia , Transducción de Señal
12.
EMBO Rep ; 5(11): 1084-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15514680

RESUMEN

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.


Asunto(s)
Apoptosis , FN-kappa B/metabolismo , Transducción de Señal , Receptor fas/biosíntesis , Alelos , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Cloranfenicol O-Acetiltransferasa/metabolismo , Ensayo de Inmunoadsorción Enzimática , Exones , Heterocigoto , Homocigoto , Humanos , Ligandos , Linfocitos/metabolismo , Ratones , Modelos Biológicos , Mutación , Plásmidos/metabolismo , Bazo/citología , Bazo/metabolismo , Factores de Tiempo
13.
Cell Cycle ; 3(10): 1235-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467462

RESUMEN

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.


Asunto(s)
FN-kappa B/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Receptor fas/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Regulación hacia Abajo/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Biológicos , Neoplasias/patología
14.
J Biol Chem ; 279(45): 46742-7, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15345718

RESUMEN

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.


Asunto(s)
Apoptosis , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Receptor fas/biosíntesis , Western Blotting , Muerte Celular , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Invasividad Neoplásica , Fosforilación , Plásmidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
15.
EMBO J ; 23(15): 3175-85, 2004 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-15272306

RESUMEN

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.


Asunto(s)
Apoptosis , Movimiento Celular , Glicoproteínas de Membrana/metabolismo , Invasividad Neoplásica/patología , Neoplasias/metabolismo , Neoplasias/patología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Caspasa 8 , Caspasas/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Proteína Ligando Fas , Humanos , Sistema de Señalización de MAP Quinasas , Glicoproteínas de Membrana/farmacología , FN-kappa B/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Receptor fas/metabolismo
16.
Oncogene ; 22(53): 8634-44, 2003 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-14634625

RESUMEN

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


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis , Proteínas de Unión al ADN , Péptidos y Proteínas de Señalización Intracelular , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/fisiología , Caspasa 10 , Caspasa 8 , Caspasa 9 , Caspasas/fisiología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Datos de Secuencia Molecular , Neoplasias/terapia , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Proteínas/fisiología
17.
Semin Immunol ; 15(3): 185-93, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-14563117

RESUMEN

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.


Asunto(s)
Mitocondrias/metabolismo , Transducción de Señal , Receptor fas/metabolismo , Animales , Proteína Ligando Fas , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Modelos Inmunológicos , Receptor fas/inmunología
18.
Proc Natl Acad Sci U S A ; 100(20): 11445-50, 2003 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-14504390

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/patología , Receptor fas/inmunología , Apoptosis , Línea Celular , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias/inmunología
19.
Cell ; 114(2): 148-50, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12887914
20.
Curr Opin Cell Biol ; 14(6): 721-6, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12473345

RESUMEN

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.


Asunto(s)
Caspasas/fisiología , Animales , Apoptosis , Ciclo Celular , División Celular , Movimiento Celular , Supervivencia Celular , Modelos Biológicos , Receptores de Superficie Celular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...