Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 2(12)2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28783662

RESUMEN

CD4+FOXP3+ regulatory T (Treg) cells are critical mediators of immune tolerance, and their deficiency owing to FOXP3 mutations in immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) patients results in severe autoimmunity. Different FOXP3 mutations result in a wide range of disease severity, reflecting the relative importance of the affected residues in the integrity of the FOXP3 protein and its various molecular interactions. We characterized the cellular and molecular impact of the most common IPEX mutation, p.A384T, on patient-derived Treg cells. We found that the p.A384T mutation abrogated the suppressive capacity of Treg cells while preserving FOXP3's ability to repress inflammatory cytokine production. This selective functional impairment is partly due to a specific disruption of FOXP3A384T binding to the histone acetyltransferase Tat-interacting protein 60 (TIP60) (KAT5) and can be corrected using allosteric modifiers that enhance FOXP3-TIP60 interaction. These findings reveal the functional impact of TIP60 in FOXP3-driven Treg biology and provide a potential target for therapeutic manipulation of Treg activity.

2.
Cell Rep ; 1(6): 665-75, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22813742

RESUMEN

FOXP3 is a key transcription factor for regulatory T cell function. We report the crystal structure of the FOXP3 coiled-coil domain, through which a loose or transient dimeric association is formed and modulated, accounting for the activity variations introduced by disease-causing mutations or posttranslational modifications. Structure-guided mutagenesis revealed that FOXP3 coiled-coil-mediated homodimerization is essential for Treg function in vitro and in vivo. In particular, we identified human FOXP3 K250 and K252 as key residues for the conformational change and stability of the FOXP3 dimer, which can be regulated by protein posttranslational modifications such as reversible lysine acetylation. These studies provide structural and mechanistic explanations for certain disease-causing mutations in the coiled-coil domain of FOXP3 that are commonly found in IPEX syndrome. Overall, the regulatory machinery involving homooligomerization, acetylation, and heteroassociation has been dissected, defining atomic insights into the biological and pathological characteristics of the FOXP3 complex.


Asunto(s)
Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/metabolismo , Multimerización de Proteína/inmunología , Linfocitos T Reguladores/inmunología , Acetilación/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Análisis Mutacional de ADN , Factores de Transcripción Forkhead/ultraestructura , Humanos , Células Jurkat , Leucina Zippers , Lisina/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Linfocitos T Reguladores/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Dedos de Zinc
3.
J Biol Chem ; 287(8): 5317-26, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22170055

RESUMEN

The SUN (Sad1-UNC-84 homology) domain is conserved in a number of nuclear envelope proteins involved in nuclear migration, meiotic telomere tethering, and antiviral responses. The LINC (linker of nucleoskeleton and cytoskeleton) complex, formed by the SUN and the nesprin proteins at the nuclear envelope, serves as a mechanical linkage across the nuclear envelope. Here we report the crystal structure of the SUN2 protein SUN domain, which reveals a homotrimer. The SUN domain is sufficient to mediate binding to the KASH (Klarsicht, ANC-1, and Syne homology) domain of nesprin 2, and the regions involved in the interaction have been identified. Binding of the SUN domain to the KASH domain is abolished by deletion of a region important for trimerization or by point mutations associated with nuclear migration failure. We propose a model of the LINC complex, where the SUN and the KASH domains form a higher ordered oligomeric network in the nuclear envelope. These findings provide the structural basis for understanding the function and the regulation of the LINC complex.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Membrana Nuclear/química , Membrana Nuclear/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Citoesqueleto/metabolismo , Células HEK293 , Humanos , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
4.
Bioorg Med Chem Lett ; 21(8): 2550-3, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21392985

RESUMEN

EGFR is over-expressed in several solid tumors including breast, prostate, pancreas, and lung cancers and is correlated to the metastatic potential of the tumor. Anti-EGFR receptor-binding peptidomimetics (AERP) were examined to assess the small molecule's potential use as tumor-specific imaging agents. The aim of this work was to design and characterize the binding specificity of the radiolabeled peptidomimetics to EGFR over-expressing cell lysate and to A431 xenograft tumors. Our newly designed peptidomimetic, AERP, was conjugated to DTPA and labeled with (99m)Tc. The in vivo tumor accumulation of [(99m)Tc] DTPA-AERP-2 was 1.6±0.1%ID/g and tumor to muscle ratio was 5.5. Our studies suggest that this novel peptidomimetic, AERP-2, warrants further development as an EGFR specific tumor-imaging agent.


Asunto(s)
Inhibidores Enzimáticos/química , Receptores ErbB/antagonistas & inhibidores , Neoplasias/diagnóstico por imagen , Radiofármacos/química , Animales , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Receptores ErbB/metabolismo , Humanos , Ratones , Ratones Desnudos , Ácido Pentético/química , Peptidomiméticos , Cintigrafía , Radiofármacos/síntesis química , Tecnecio/química , Distribución Tisular , Trasplante Heterólogo
5.
Semin Cell Dev Biol ; 21(9): 961-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20850557

RESUMEN

Our work is concerned with the origins and therapy of human cancers. Members of the epidermal growth factor receptor (EGFR) family of tyrosine kinases, also known as erbB or HER receptors, are over expressed and/or activated in many types of human tumors and represent important therapeutic targets in cancer therapy. Studies from our laboratory identified targeted therapy as a way to treat cancer. Rational therapeutics targeting and disabling erbB receptors have been developed to reverse the malignant properties of tumors. Reversal of the malignant phenotype, best seen with disabling the HER2 receptors using monoclonal antibodies is a distinct process from that seen with blocking of ligand binding to cognate receptors as has been done for EGFr receptors. Here we review the mechanisms of action deduced from a number of approaches developed in our laboratory and elsewhere, including monoclonal antibodies, peptide mimetics, recombinant proteins and small molecules. The biochemical and biological principles which have been uncovered during these studies of disabling HER2 homomeric or HER2-EGFr heteromeric receptors will help the development of novel and more efficient therapeutics targeting erbB family receptors.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Humanos , Neoplasias/inmunología
6.
Int Immunopharmacol ; 9(5): 518-20, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19539569

RESUMEN

The forkhead family transcription factor FOXP3 plays a fundamental role in immune homeostasis. FOXP3 dysfunction in regulatory T cells (Tregs) contributes to multiple disease processes such as autoimmunity, tumor development, and viral infection. FOXP3 cooperates and associates with a group of other transcriptional factors, co-repressors and co-activators in Tregs to form one or more dynamic regulatory complexes. These ensembles communicate with multiple key signaling pathways to either upregulate or downregulate the expression of downstream target genes such as cytokines and cell surface receptors, which are critical for the control of normal immune responses. Although the details of the underlying mechanism by which FOXP3 operates as a transcriptional repressor or an activator is largely undefined, FOXP3(+) Tregs based cellular therapies have been studied in animal models. Our recent studies concerning the FOXP3 complex ensemble provide structural and biochemical insights into FOXP3 function of Tregs, which are essential to the development of novel immunopharmacological agents for treating human immunological disease.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Sitios de Unión , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dimerización , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Linfocitos T Reguladores/inmunología
7.
Curr Opin Investig Drugs ; 9(12): 1336-46, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19037840

RESUMEN

The anilino-quinazoline derivative BIBW-2992, which is being developed by Boehringer Ingelheim Corp for the potential treatment of solid tumors, is an oral dual receptor tyrosine kinase inhibitor of human EGF receptor (EGFR) and human epidermal growth factor receptor-2 (HER-2)/neu. EGFR and HER-2/neu activate numerous signaling pathways leading to cancer cell proliferation, survival and migration. In vitro, BIBW-2992 effectively and selectively inhibited EGFR and HER-2/neu and inhibited EGFR and HER-2/neu total tyrosine phosphorylation and tumor cell proliferation in vivo. Importantly, BIBW-2992 was active against tumors overexpressing EGFR with the secondary Thr790Met point mutation, which confers resistance to the first-generation EGFR inhibitors gefitinib and erlotinib. In phase I/II trials, BIBW-2992 was effective in patients with solid tumors, including those with NSCLC tumors activating mutations in the EGFR tyrosine kinase domain. BIBW-2992 was generally well tolerated with the main adverse effects being gastrointestinal or cutaneous disorders. At the time of publication, BIBW-2992 was undergoing phase II trials for NSCLC, breast and prostate cancers, head and neck carcinoma, as well as glioma. BIBW-2992 was granted Fast-Track status by the FDA for NSCLC and was investigated in phase III trials for this indication.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Quinazolinas/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Afatinib , Animales , Ensayos Clínicos como Asunto , Ensayos de Selección de Medicamentos Antitumorales , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Humanos , Quinazolinas/farmacología
8.
Methods ; 46(1): 39-46, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18571508

RESUMEN

Although numerous biomolecular interactions have been identified as potential targets for the development of therapeutic agents, modulation of these interactions with small molecules has historically been considered an extremely difficult task. This difficulty is largely due to the low effectiveness of the traditionally used competitive approaches in which inhibitors are designed and screened for their ability to block biomolecules from establishing contacts with their targets. We propose a novel approach to modulate biomolecular interactions by de novo structure-based design of noncompetitive small molecules that bind to the intermolecular complexes and make molecular contacts with both interacting partners. Similar complex-binding mechanism has been observed and well documented for many natural compounds that bind to protein-protein, protein-DNA or protein-small molecule complexes. To implement the paradigm for structure-based drug design, we have developed a complex-binding modulation (CBM) algorithm for the rational design of compounds (CBM compounds) that can affect biomolecular interactions by binding to the intermolecular pockets or cavities of biomolecular complexes. In this paper, we describe our methodology used to design the CBM compounds and to study their effects on biomolecular interactions including protein-protein and protein-small molecule interactions.


Asunto(s)
Ligandos , Sustancias Macromoleculares/química , Mapeo de Interacción de Proteínas/tendencias , Sitios de Unión , Biofisica/tendencias , Diseño de Fármacos , Nanotecnología/tendencias , Unión Proteica , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
9.
Proc Natl Acad Sci U S A ; 105(14): 5465-70, 2008 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-18378892

RESUMEN

Ligation between glucocorticoid-induced tumor necrosis factor receptor (GITR) and its ligand (GITRL) provides an undefined signal that renders CD4(+)CD25(-) effector T cells resistant to the inhibitory effects of CD4(+)CD25(+) regulatory T cells. To understand the structural basis of GITRL function, we have expressed and purified the extracellular domain of human GITR ligand in Escherichia coli. Chromotography and cross-linking studies indicate that human GITRL (hGITRL) exists as dimers and trimers in solution and also can form a supercluster. To gain insight into the nature of GITRL oligomerization, we determined the crystallographic structures of hGITRL, which revealed a loosely associated open trimer with a deep cavity at the molecular center and a flexible C-terminal tail bent for trimerization. Moreover, a tetramer of trimers (i.e., supercluster) has also been observed in the crystal, consistent with the cross-linking analysis. Deletion of the C-terminal distal three residues disrupts the loosely assembled trimer and favors the formation of a dimer that has compromised receptor binding and signaling activity. Collectively, our studies identify multiple oligomeric species of hGITRL that possess distinct kinetics of ERK activation. The studies address the functional implications and structural models for a process by which hGITRL utilizes multiple oligomerization states to regulate GITR-mediated signaling during T cell costimulation.


Asunto(s)
Factores de Necrosis Tumoral/química , Clonación Molecular , Cristalografía por Rayos X , Dimerización , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Humanos , Cinética , Conformación Proteica , Estructura Cuaternaria de Proteína , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Soluciones , Factores de Necrosis Tumoral/metabolismo
10.
J Clin Invest ; 117(8): 2051-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17671639

RESUMEN

Understanding the genetic origin of cancer at the molecular level has facilitated the development of novel targeted therapies. Aberrant activation of the ErbB family of receptors is implicated in many human cancers and is already the target of several anticancer therapeutics. The use of mAbs specific for the extracellular domain of ErbB receptors was the first implementation of rational targeted therapy. The cytoplasmic tyrosine kinase domain is also a preferred target for small compounds that inhibit the kinase activity of these receptors. However, current therapy has not yet been optimized, allowing for opportunities for optimization of the next generation of targeted therapy, particularly with regards to inhibiting heteromeric ErbB family receptor complexes.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Animales , Sistemas de Liberación de Medicamentos , Activación Enzimática/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias/genética , Estructura Terciaria de Proteína
11.
J Immunol ; 178(2): 1021-9, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17202365

RESUMEN

mAbs capable of disabling heterodimeric kinase complexes of the epidermal growth factor receptor (EGFR) and human EGFR type 2/neu have therapeutic relevance to various human cancers. In this study, we demonstrate that in addition to the dimer, EGFR and human EGFR type 2 can associate as homo- and heterotetramers. EGF-induced phosphorylation of the tetramers was significantly lower than that of the dimers, indicating that the tetrameric receptor complexes have impaired signaling activity. Targeting v-erb-b2 erythroblastic leukemia viral oncogene homolog (erbB) receptors with mAbs promoted erbB tetrameric assembly, suggesting that a component of the antitumor activity may be mediated by the ability of Abs to shift the equilibrium from active dimeric to impaired tetrameric receptor complex states. This study suggests a novel therapeutic approach to disable signaling of erbB and potentially other receptors in tumors by biologic agents capable of inducing receptor tetramerization.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Animales , Catálisis , Línea Celular , Chlorocebus aethiops , Receptores ErbB/metabolismo , Eliminación de Gen , Humanos , Peso Molecular , Unión Proteica , Receptor ErbB-2/genética
13.
Protein J ; 24(7-8): 465-78, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16328739

RESUMEN

The interaction of the precursor to mitochondrial aspartate aminotransferase (pmAAT) with GroEL has been studied by electron paramagnetic resonance (EPR) and fluorescence spectroscopy. In the native protein, the spin probe was immobilized when attached to Cys166 at the domain interface, but was fully mobile when introduced at Cys(-19) in the N-terminal presequence peptide. Unfolding of the protein resulted in a highly mobile EPR spectrum for probes introduced at either site. However, the nitroxide group in GroEL-bound pmAAT showed either intermediate or high mobility depending on the spin probe used. Power saturation experiments indicated that the accessibility of the nitroxide side chain to Ni(EDDA) in the GroEL-pmAAT complex was higher than in the native state when in position 166 but lower when at position -19. Similar results were obtained in fluorescence quenching experiments. These data suggest that GroEL binds partly folded states of pmAAT with the presequence peptide probably in direct contact with GroEL. GroES and ATP, but not AMP-PNP or ADP, support refolding of pmAAT. During refolding, the rate of recovery of the native spectroscopic properties of labeled Cys166 is nearly identical to the rate-limiting reactivation step. Thus, correct docking of the large and small domains of pmAAT may be a key structural event in the regain of catalytic activity.


Asunto(s)
Aspartato Aminotransferasas/química , Chaperonina 60/química , Espectroscopía de Resonancia por Spin del Electrón , Conformación Proteica , Aspartato Aminotransferasas/metabolismo , Chaperonina 60/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Modelos Moleculares , Estructura Molecular , Unión Proteica , Desnaturalización Proteica , Pliegue de Proteína , Solventes/química , Espectrometría de Fluorescencia , Marcadores de Spin
14.
Proc Natl Acad Sci U S A ; 102(31): 10970-5, 2005 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-16043718

RESUMEN

We have disabled TNF receptor (TNFR) function by inducing allosteric modulation of tryptophan-107 (W107) in the receptor. The allosteric effect operates by means of an allosteric cavity found a short distance from a previously identified loop involved in ligand binding. Occupying this cavity by small molecules leads to perturbation of distal W107 and disables functions of the TNFR, a molecule not known to undergo conformational change upon binding TNF-alpha. TNF-alpha-induced NF-kappaB and p38 kinase activities and clinical symptoms of collagen-induced arthritis in mice were all diminished. Thus, disabling receptor function by induced conformational changes of active binding surfaces represents an innovative paradigm in structure-based drug design.


Asunto(s)
Receptores Tipo I de Factores de Necrosis Tumoral/química , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Sitio Alostérico/genética , Sustitución de Aminoácidos , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Experimental/prevención & control , Secuencia de Bases , Línea Celular , ADN/genética , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos DBA , Modelos Moleculares , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , Conformación Proteica , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal , Triptófano/química , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
DNA Cell Biol ; 24(6): 350-8, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15941387

RESUMEN

HER2 is a validated therapeutic target for cancer. There are no natural ligands, but monoclonal antibodies and peptides that bind HER2 act as artificial ligands, selectively affecting HER2-overexpressing tumors. One reported mechanism for this effect is receptor downregulation, but the expected correlation of ligand-dependent HER2 internalization and tumor inhibition remain poorly characterized. Moreover, HER2 ligands have limited therapeutic efficacy and often they require adjuvant treatment with the chemotherapeutic Taxol. Here, we generated a series of HER2 ligands (Anti-HER2/neu peptide ligands, AHNPmonovalent and AHNPbivalent) with different valency and correlated their internalization-promoting ability to biological potency. Since AHNPbivalent (but not AHNPmonovalent) induces rapid receptor internalization, we exploited this feature to deliver cytotoxic conjugates coupling AHNPbivalent and Taxol (Taxol . AHNPbivalent). The prodrug conjugate releases Taxol after receptor-mediated internalization, and cytotoxicity can be used as a marker of internalization. Taxol . AHNPbivalent is significantly more cytotoxic than free Taxol + free AHNPbivalent. Hence, the Taxol x AHNP(bivalent) prodrug binds to HER2, induces receptor internalization and downregulation, and the subsequent release of free Taxol inside the targeted cell results in synergistic toxicity, The effect is selective towards HER2- expressing cells. This work links HER2 receptor internalization and growth arrest, and the chemical conjugation strategy may yield improved and HER2 selective therapeutics.


Asunto(s)
Endocitosis/efectos de los fármacos , Inmunotoxinas/farmacología , Profármacos/farmacología , Receptor ErbB-2/metabolismo , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Unión Competitiva/inmunología , Supervivencia Celular/efectos de los fármacos , Dimerización , Relación Dosis-Respuesta a Droga , Inmunotoxinas/química , Ratones , Modelos Moleculares , Estructura Molecular , Células 3T3 NIH , Paclitaxel/química , Paclitaxel/farmacología , Profármacos/química , Conformación Proteica , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Transfección
16.
DNA Cell Biol ; 23(8): 475-89, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15307950

RESUMEN

The centrosome is a unique organelle that functions as the microtubule organizing center in most animal cells. During cell division, the centrosomes form the poles of the bipolar mitotic spindle. In addition, the centrosomes are also needed for cytokinesis. Each mammalian somatic cell typically contains one centrosome, which is duplicated in coordination with DNA replication. Just like the chromosomes, the centrosome is precisely reproduced once and only once during each cell cycle. However, it remains a mystery how this protein-based structure undergoes accurate duplication in a semiconservative manner. Intriguingly, amplification of the centrosome has been found in numerous forms of cancers. Cells with multiple centrosomes tend to form multipolar spindles, which result in abnormal chromosome segregation during mitosis. It has therefore been postulated that centrosome aberration may compromise the fidelity of cell division and cause chromosome instability. Here we review the current understanding of how the centrosome is assembled and duplicated. We also discuss the possible mechanisms by which centrosome abnormality contributes to the development of malignant phenotype.


Asunto(s)
Centrosoma/fisiología , Mitosis/fisiología , Modelos Biológicos , Huso Acromático/fisiología , Aneuploidia , Aurora Quinasas , Quinasas CDC2-CDC28/fisiología , Proteínas de Ciclo Celular , División Celular/fisiología , Transformación Celular Neoplásica/patología , Inestabilidad Cromosómica/fisiología , Quinasa 2 Dependiente de la Ciclina , Replicación del ADN/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Fosfatasas , Proteínas de Saccharomyces cerevisiae
17.
Proc Natl Acad Sci U S A ; 101(17): 6599-604, 2004 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15084739

RESUMEN

Fas ligand- (FasL) mediated apoptosis is an important element of tissue-specific organ damage. We have developed biologically active small exocyclic peptide mimetics that disable apoptotic functions of Fas. The most effective mimetic binds to both its receptor and FasL with comparable affinity. In vitro, the most effective antagonist blocked FasL-induced cytotoxicity completely and specifically. In vivo, the antagonistic mimetic also prevented Concanavilin A (Con A) induced hepatitis, a CD4(+) T cell-mediated animal model of liver injury. Although current approaches prevent Fas receptor signaling by excluding FasL binding to Fas, the small molecule mimetics reported here disable Fas by promoting a defective Fas-FasL receptor complex. This event desensitizes FasL-mediated apoptosis by inhibiting extracellular signal regulated kinase activity and up-regulating NF-kappaB.


Asunto(s)
Apoptosis/efectos de los fármacos , Glicoproteínas de Membrana/antagonistas & inhibidores , Imitación Molecular , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Western Blotting , Concanavalina A/farmacología , Proteína Ligando Fas , Citometría de Flujo , Hígado/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Moleculares , FN-kappa B/metabolismo , Péptidos/química , Péptidos/metabolismo , Unión Proteica
18.
Immunol Res ; 27(2-3): 303-8, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12857977

RESUMEN

The epidermal growth factor (EGF) family of tyrosine kinase receptors (erbB receptors) are expressed at high levels in a wide variety of human cancers and have been associated with various features of advanced disease and poor prognosis. Therapeutic blockade of erbB signaling is a novel approach to the treatment of human tumors that could offer a noncytotoxic alternative to cancer treatment. A number of monoclonal antibodies (MAbs) directed against erbB receptors have been developed and demonstrated promising therapeutic results. We have designed small-molecule peptide mimetics of an anti-erbB rhu MAb 4D5 that can mimic structural and functional properties of the parental antibody. An alternative structure-based strategy of erbB receptor blockade with peptide mimetics by targeting receptor dimerization interfaces is also described.


Asunto(s)
Anticuerpos Monoclonales/química , Péptidos/inmunología , Receptor ErbB-2/antagonistas & inhibidores , Biomimética , Dimerización , Relación Estructura-Actividad
19.
Exp Mol Pathol ; 73(2): 93-103, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12231211

RESUMEN

We designed a new class of aromatically modified exocyclic peptides based on the structure of CD4 by engineering one of the cysteine residues in a peptidomimetic derived from the CDR3 region of the CD4 molecule. All three species mediate inhibition of T-cell proliferation at concentrations ranging from 10 to 100 microM. The mimetics CD4-Cys and CD4-Met bind to sCD4 with affinities ranging from 1 to 2 microM, while CD4-Ser shows poor binding in radioisotope assay. Though these mimetics have similar structures, they exhibit different biochemical and biological functions. Activation of T-cells as measured by thymidine incorporation or IL-2 production revealed that CD4-Cys and CD4-Ser mimetics behave as classical antagonists. On the other hand, the CD4-Met species inhibited T-cell proliferation with an IC(50) of 30 microM but unexpectedly increased IL-2 secretion modestly at a less than 3 microM concentration. In experimental autoimmune encephalitis (EAE), CD4-Ser and CD4-Cys mimetics reduced the severity of EAE symptoms while the CD4-Met mimetic exacerbated the conditions. We propose that CD4-Cys and CD4-Ser are classical antagonists, but CD4-Met may possess properties of an inverse agonist. The structure-activity relationship of mimetics reveals that a minor change in the net hydropathic value is enough to alter the dynamic nature of the receptor-ligand complex.


Asunto(s)
Antígenos CD4/química , Antígenos CD4/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/inmunología , Animales , Biotecnología , Antígenos CD4/farmacología , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/metabolismo , Simulación por Computador , Dimerización , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Cobayas , Humanos , Técnicas In Vitro , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos , Modelos Biológicos , Modelos Moleculares , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Conformación Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos
20.
J Biol Chem ; 277(31): 28330-9, 2002 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-12011054

RESUMEN

Members of the erbB family receptor tyrosine kinases (erbB1, erbB2, erbB3, and erbB4) are overexpressed in a variety of human cancers and represent important targets for the structure-based drug design. Homo- and heterodimerization (oligomerization) of the erbB receptors are known to be critical events for receptor signaling. To block receptor self-associations, we have designed a series of peptides derived from potential dimerization surfaces in the extracellular subdomain IV of the erbB receptors (erbB peptides). In surface plasmon resonance (BIAcore) studies, the designed peptides have been shown to selectively bind to the erbB receptor ectodomains and isolated subdomain IV of erbB2 with submicromolar affinities and to inhibit heregulin-induced interactions of erbB3 with different erbB receptors. A dose-dependent inhibition of native erbB receptor dimerization by the erbB peptides has been observed in 32D cell lines transfected with different combinations of erbB receptors. The peptides effectively inhibited growth of two types of transformed cells overexpressing different erbB receptors, T6-17 and 32D, in standard MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and cell viability assays. The study identifies distinct loops within the membrane-proximal part of the subdomain IV as potential receptor-receptor interaction sites for the erbB receptors and demonstrates the possibility of disabling receptor activity by structure-based targeting of the dimerization interfaces. Molecular models for possible arrangement of the erbB1.EGF complex, consistent with the involvement of subdomain IV in inter-receptor interactions, are proposed. Small dimerization inhibitors described herein can be useful as probes to elucidate different erbB signaling pathways and may be developed as therapeutic agents.


Asunto(s)
Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/metabolismo , Secuencia de Aminoácidos , Dimerización , Diseño de Fármacos , Receptores ErbB/química , Receptores ErbB/metabolismo , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Conformación Proteica , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA