Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 163(6): 1333-47, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26607792

RESUMEN

Interphase chromatin is organized in distinct nuclear sub-compartments, reflecting its degree of compaction and transcriptional status. In Caenorhabditis elegans embryos, H3K9 methylation is necessary to silence and to anchor repeat-rich heterochromatin at the nuclear periphery. In a screen for perinuclear anchors of heterochromatin, we identified a previously uncharacterized C. elegans chromodomain protein, CEC-4. CEC-4 binds preferentially mono-, di-, or tri-methylated H3K9 and localizes at the nuclear envelope independently of H3K9 methylation and nuclear lamin. CEC-4 is necessary for endogenous heterochromatin anchoring, but not for transcriptional repression, in contrast to other known H3K9 methyl-binders in worms, which mediate gene repression but not perinuclear anchoring. When we ectopically induce a muscle differentiation program in embryos, cec-4 mutants fail to commit fully to muscle cell fate. This suggests that perinuclear sequestration of chromatin during development helps restrict cell differentiation programs by stabilizing commitment to a specific cell fate. PAPERCLIP.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Proteínas Cromosómicas no Histona/metabolismo , Embrión no Mamífero/citología , Heterocromatina , Código de Histonas , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Diferenciación Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Embrión no Mamífero/metabolismo , Datos de Secuencia Molecular , Alineación de Secuencia
2.
Blood ; 142(12): 1071-1081, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37294924

RESUMEN

Rebalance of coagulation and anticoagulation to achieve a hemostatic effect has recently gained attention as an alternative therapeutic strategy for hemophilia. We engineered a humanized chimeric antibody, SR604, based on a previously published murine antibody, HAPC1573, which selectively blocks the anticoagulant activity of human activated protein C (APC). SR604 effectively blocked the anticoagulation activities of APC in human plasma deficient in various coagulation factors in vitro with affinities ∼60 times greater than that of HAPC1573. SR604 exhibited prophylactic and therapeutic efficacy in the tail-bleeding and knee-injury models of hemophilia A and B mice expressing human APC (humanized hemophilic mice). SR604 did not interfere with the cytoprotection and endothelial barrier function of APC, nor were there obvious toxicity effects in humanized hemophilic mice. Pharmacokinetic study showed a high bioavailability (106%) of subcutaneously injected SR604 in cynomolgus monkeys. These results demonstrate that SR604 is expected to be a safe and effective therapeutic and/or prophylactic agent with a prolonged half-life for patients with congenital factor deficiencies including hemophilia A and B.


Asunto(s)
Hemofilia A , Proteína C , Humanos , Ratones , Animales , Proteína C/uso terapéutico , Hemofilia A/tratamiento farmacológico , Modelos Animales de Enfermedad , Coagulación Sanguínea , Anticoagulantes/uso terapéutico
3.
Neuropathol Appl Neurobiol ; 48(5): e12819, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35508761

RESUMEN

AIM: Tau truncation (tr-tau) by active caspase-6 (aCasp-6) generates tau fragments that may be toxic. Yet the relationship between aCasp-6, different forms of tr-tau and hyperphosphorylated tau (p-tau) accumulation in human brains with Alzheimer's disease (AD) and other tauopathies remains unclear. METHODS: We generated two neoepitope monoclonal antibodies against tr-tau sites (D402 and D13) targeted by aCasp-6. Then, we used five-plex immunofluorescence to quantify the neuronal and astroglial burden of aCasp-6, tr-tau, p-tau and their co-occurrence in healthy controls, AD and primary tauopathies. RESULTS: Casp-6 activation was strongest in AD and Pick's disease (PiD) but almost absent in 4-repeat (4R) tauopathies. In neurons, the tr-tau burden was much more abundant in AD and PiD than in 4R tauopathies and disproportionally higher when normalising by p-tau pathology. Tr-tau astrogliopathy was detected in low numbers in 4R tauopathies. Unexpectedly, about half of tr-tau positive neurons in AD and PiD lacked p-tau aggregates, a finding we confirmed using several p-tau antibodies. CONCLUSIONS: Early modulation of aCasp-6 to reduce tr-tau pathology is a promising therapeutic strategy for AD and PiD but is unlikely to benefit 4R tauopathies. The large percentage of tr-tau-positive neurons lacking p-tau suggests that many vulnerable neurons to tau pathology go undetected when using conventional p-tau antibodies. Therapeutic strategies against tr-tau pathology could be necessary to modulate the extent of tau abnormalities in AD. The disproportionally higher burden of tr-tau in AD and PiD supports the development of biofluid biomarkers against tr-tau to detect AD and PiD and differentiate them from 4R tauopathies at a patient level.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Encéfalo/patología , Caspasa 6 , Humanos , Neuronas/patología , Tauopatías/diagnóstico , Tauopatías/patología , Tauopatías/terapia , Proteínas tau/metabolismo
4.
Adv Exp Med Biol ; 1248: 619-649, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32185726

RESUMEN

Immune checkpoint blockade (ICB) has been proven to be an effective strategy for enhancing the effector activity of anti-tumor T cells, and checkpoint blockers targeting CTLA-4, PD-1, and PD-L1 have displayed strong and durable clinical responses in certain cancer patients. The new hope brought by ICB therapy has led to the boost in therapeutic development of ICBs in recent years. Nonetheless, the therapeutic efficacy of ICBs varies substantially among cancer types and patients, and only a proportion of cancer patients could benefit from ICBs. The emerging targets and molecules for enhancing anticancer immunity may bring additional therapeutic opportunities for cancer patients. The current challenges in the ICB therapy have been discussed, aimed to provide further strategies for maximizing the efficacy of ICB therapy.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Humanos , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
5.
Proc Natl Acad Sci U S A ; 109(52): 21360-5, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236167

RESUMEN

Ezh2 (Enhancer of zeste homolog 2) protein is the enzymatic component of the Polycomb repressive complex 2 (PRC2), which represses gene expression by methylating lysine 27 of histone H3 (H3K27) and regulates cell proliferation and differentiation during embryonic development. Recently, hot-spot mutations of Ezh2 were identified in diffused large B-cell lymphomas and follicular lymphomas. To investigate if tumor growth is dependent on the enzymatic activity of Ezh2, we developed a potent and selective small molecule inhibitor, EI1, which inhibits the enzymatic activity of Ezh2 through direct binding to the enzyme and competing with the methyl group donor S-Adenosyl methionine. EI1-treated cells exhibit genome-wide loss of H3K27 methylation and activation of PRC2 target genes. Furthermore, inhibition of Ezh2 by EI1 in diffused large B-cell lymphomas cells carrying the Y641 mutations results in decreased proliferation, cell cycle arrest, and apoptosis. These results provide strong validation of Ezh2 as a potential therapeutic target for the treatment of cancer.


Asunto(s)
Linfoma de Células B Grandes Difuso/patología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Embrión de Mamíferos/citología , Proteína Potenciadora del Homólogo Zeste 2 , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Linfoma de Células B Grandes Difuso/genética , Metilación/efectos de los fármacos , Ratones , Mutación/genética , Fenotipo , Complejo Represivo Polycomb 2/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
6.
J Biol Chem ; 288(5): 3477-88, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23258544

RESUMEN

Insulin resistance, hyperlipidemia, and cardiovascular complications are common dysregulations of metabolic syndrome. Transplant patients treated with immunosuppressant drugs such as cyclosporine A (CsA), an inhibitor of calcineurin phosphatase, frequently develop similar metabolic complications. Although calcineurin is known to mediate insulin sensitivity by regulating ß-cell growth and adipokine gene transcription, its role in lipid homeostasis is poorly understood. Here, we examined lipid homeostasis in mice lacking calcineurin Aß (CnAß(-/-)). We show that mice lacking calcineurin Aß are hyperlipidemic and develop age-dependent insulin resistance. Hyperlipidemia found in CnAß(-/-) mice is, in part, due to increased lipolysis in adipose tissues, a process mediated by ß-adrenergic G-protein-coupled receptor signaling pathways. CnAß(-/-) mice also exhibit additional pathophysiological phenotypes caused by the potentiated GPCR signaling pathways. A cell autonomous mechanism with sustained cAMP/PKA activation is found in CnAß(-/-) mice or upon CsA treatment to inhibit calcineurin. Increased PKA activation and cAMP accumulation in CnAß(-/-) mice, however, are sensitive to phosphodiesterase inhibitor. Indeed, we show that calcineurin regulates degradation of phosphodiesterase 3B, in addition to phosphodiesterase 4D. These results establish a role for calcineurin in lipid homeostasis. These data also indicate that potentiated cAMP signaling pathway may provide an alternative molecular pathogenesis for the metabolic complications elicited by CsA in transplant patients.


Asunto(s)
Calcineurina/deficiencia , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Hiperlipidemias/enzimología , Transducción de Señal , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Secuencia de Aminoácidos , Animales , Células COS , Calcineurina/metabolismo , Chlorocebus aethiops , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Ciclosporina/farmacología , Embrión de Mamíferos/citología , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Hiperlipidemias/patología , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Inhibidores de Fosfodiesterasa/farmacología , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/efectos de los fármacos , Triglicéridos/biosíntesis
7.
FEBS Open Bio ; 13(7): 1253-1265, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37302810

RESUMEN

Lymphocyte activation gene-3 (LAG-3) is a type I transmembrane protein with structural similarities to CD4. Overexpression of LAG-3 enables cancer cells to escape immune surveillance, while its blockade reinvigorates exhausted T cells and strengthens anti-infection immunity. Blockade of LAG-3 may have antitumor effects. Here, we generated a novel anti-LAG-3 chimeric antibody, 405B8H3(D-E), through hybridoma technology from monoclonal antibodies produced in mice. The heavy-chain variable region of the selected mouse antibody was grafted onto a human IgG4 scaffold, while a modified light-chain variable region was coupled to the human kappa light-chain constant region. 405B8H3(D-E) could effectively bind LAG-3-expressing HEK293 cells. Moreover, it could bind cynomolgus monkey (cyno) LAG-3 expressed on HEK293 cells with a higher affinity than the reference anti-LAG-3 antibody BMS-986016. Furthermore, 405B8H3(D-E) promoted interleukin-2 secretion and was able to block the interactions of LAG-3 with liver sinusoidal endothelial cell lectin and major histocompatibility complex II molecules. Finally, 405B8H3(D-E) combined with anti-mPD-1-antibody showed effective therapeutic potential in the MC38 tumor mouse model. Therefore, 405B8H3(D-E) is likely to be a promising candidate therapeutic antibody for immunotherapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales , Ratones , Humanos , Animales , Células HEK293 , Macaca fascicularis/metabolismo
8.
MAbs ; 15(1): 2184197, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36859773

RESUMEN

The urokinase-type plasminogen activator receptor (uPAR) is an essential regulator for cell signaling in tumor cell proliferation, adhesion, and metastasis. The ubiquitous nature of uPAR in many aggressive cancer types makes uPAR an attractive target for immunotherapy. Here, we present a rapid and successful workflow for developing cross-reactive anti-uPAR recombinant antibodies (rAbs) using high-throughput optofluidic screening of single B-cells from human uPAR-immunized mice. A total of 80 human and cynomolgus uPAR cross-reactive plasma cells were identified, and selected mouse VH/VL domains were linked to the trastuzumab (Herceptin®) constant domains for the expression of mouse-human chimeric antibodies. The resulting rAbs were characterized by their tumor-cell recognition, binding activity, and cell adhesion inhibition on triple-negative breast cancer cells. In addition, the rAbs were shown to enact antibody-dependent cellular cytotoxicity (ADCC) in the presence of either human natural killer cells or peripheral blood mononuclear cells, and were evaluated for the potential use of uPAR-targeting antibody-drug conjugates (ADCs). Three lead antibodies (11857, 8163, and 3159) were evaluated for their therapeutic efficacy in vivo and were shown to suppress tumor growth. Finally, the binding epitopes of the lead antibodies were characterized, providing information on their unique binding modes to uPAR. Altogether, the strategy identified unique cross-reactive antibodies with ADCC, ADC, and functional inhibitory effects by targeting cell-surface uPAR, that can be tested in safety studies and serve as potential immunotherapeutics.


Asunto(s)
Leucocitos Mononucleares , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Humanos , Animales , Ratones , Anticuerpos , Transducción de Señal , Linfocitos B
9.
FEBS Open Bio ; 12(8): 1542-1557, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35674216

RESUMEN

Glucocorticoid-induced TNF receptor-related (GITR) can act as a co-stimulatory receptor, representing a potential target for safely enhancing immunotherapy efficacy. GITR is triggered by a GITR ligand or an agonist antibody and activates CD8+ and CD4+ effector T cells, reducing tumor-infiltrating Treg numbers and resulting in activation of immune responses and tumor cell destruction by effector T cells. GITR is an attractive target for immunotherapy, especially in combination therapy with immune checkpoint inhibitors, as is being explored in clinical trials. Using H2L2 transgenic mice encoding the human immunoglobulin variable region and hybridoma technology, we generated a panel of fully human antibodies that showed excellent specific affinity and strong activation of human T cells. After conversion to fully human antibodies and engineering modification, we obtained an anti-GITR antibody hab019e2 with enhanced antitumor activity in a B-hGITR MC38 mouse model compared to Tab9H6V3, an anti-GITR antibody that activates T cells and inhibits Treg suppression from XenoMouse. As a fully human antibody with its posttranslational modification hot spot removed, the hab019e2 antibody exerted more potent therapeutic effects, and may have potential as a novel and developable antibody targeting GITR for follow-up drug studies.


Asunto(s)
Glucocorticoides , Receptores del Factor de Necrosis Tumoral , Animales , Anticuerpos , Línea Celular Tumoral , Proteína Relacionada con TNFR Inducida por Glucocorticoide/agonistas , Humanos , Inmunoterapia/métodos , Ratones , Receptores del Factor de Necrosis Tumoral/agonistas
10.
FEBS Open Bio ; 12(12): 2166-2178, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36176235

RESUMEN

CD137 (4-1BB, TNFRSF9), an inducible T-cell costimulatory receptor, is expressed on activated T cells, activated NK cells, Treg cells, and several innate immune cells, including DCs, monocytes, neutrophils, mast cells, and eosinophils. In animal models and clinical trials, anti-CD137 agonistic monoclonal antibodies have shown anti-tumor potential, but balancing the efficacy and toxicity of anti-CD137 agonistic monoclonal antibodies is a considerable hindrance for clinical applications. Here, we describe a novel fully human CD137 agonistic antibody (PE0116) generated from immunized harbor H2L2 human transgenic mice. PE0116 is a ligand blocker, which is also the case for Utomilumab (one of the leading CD137 agonistic drugs); PE0116 partially overlaps with Urelumab's recognized epitope. In vitro, PE0116 activates NF-κB signaling, significantly promotes T-cell proliferation, and increases cytokine secretion in the presence of cross-linking. Importantly, PE0116 possesses robust anti-tumor activity in the MC38 tumor model. In vivo, PE0116 exhibits a good safety profile and has typical pharmacokinetic characteristics of an IgG antibody in preclinical studies of non-human primates. In summary, PE0116 is a promising anti-CD137 antibody with a good safety profile in preclinical studies.


Asunto(s)
Neoplasias , Primates , Animales , Ratones , Humanos , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Células Asesinas Naturales , Linfocitos T Reguladores
11.
J Exp Med ; 219(9)2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-35881112

RESUMEN

Disease relapse and treatment-induced immunotoxicity pose significant clinical challenges for patients with hematological cancers. Here, we reveal distinctive requirements for neutralizing TNF receptor ligands APRIL and BAFF and their receptor activity in MM and DLBCL, impacting protein translation and production in MM cells and modulating the translation efficiency of the ATM interactor (ATMIN/ACSIZ). Therapeutically, we investigated the use of BCMA decoy receptor (sBCMA-Fc) as an inhibitor of APRIL and BAFF. While wild-type sBCMA-Fc effectively blocked APRIL signaling in MM, it lacked activity in DLBCL due to its weak BAFF binding. To expand the therapeutic utility of sBCMA-Fc, we engineered an affinity-enhanced mutant sBCMA-Fc fusion molecule (sBCMA-Fc V3) 4- and 500-fold stronger in binding to APRIL and BAFF, respectively. The mutant sBCMA-Fc V3 clone significantly enhanced antitumor activity against both MM and DLBCL. Importantly, we also demonstrated an adequate toxicity profile and on-target mechanism of action in nonhuman primate studies.


Asunto(s)
Linfoma de Células B Grandes Difuso , Mieloma Múltiple , Animales , Factor Activador de Células B/genética , Antígeno de Maduración de Linfocitos B/genética , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/terapia , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Transducción de Señal , Proteína Activadora Transmembrana y Interactiva del CAML , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
12.
FEBS Open Bio ; 11(3): 813-825, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33449453

RESUMEN

Cluster of differentiation 47 (CD47) is a widely expressed self-protection transmembrane protein that functions as a critical negative regulator to induce macrophage-mediated phagocytosis. Overexpression of CD47 enables cancer cells to escape immune surveillance and destruction by phagocytes both in solid tumours and leukaemia. The usefulness of anti-CD47 antibody has been demonstrated in multiple immunotherapies associated with macrophages. However, antigen sinks and toxicity induced by inadvertent binding to normal cells restrict its clinical applications. Here, a novel anti-human CD47 antibody, 4D10, was generated, and its variable regions were grafted onto a human IgG4 scaffold. Compared with the anti-CD47 antibody Hu5F9, the resulting chimeric antibody (c4D10) has consistently demonstrated good tolerance in in vitro and in vivo toxicity studies. Additionally, c4D10 showed effective therapeutic potential through inducing the eradication of human cancer cells. Thus, c4D10 is a promising candidate therapeutic antibody with higher efficacy and reduced side effects compared to earlier antibodies, and its use may reduce the dose-limiting toxicity of CD47 antagonists for immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antígeno CD47/inmunología , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Leucemia Mieloide Aguda/inmunología , Ratones , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
FEBS Lett ; 595(11): 1587-1603, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33792041

RESUMEN

OX40 is a costimulatory molecule that belongs to the tumor necrosis factor receptor (TNFR) superfamily. OX40 agonist-based combinations are emerging as promising candidates for novel cancer immunotherapy. Clinical trials have shown that OX40 agonist antibodies could lead to better results in cancer patients. Using a hybridoma platform and three different types of immunization strategies, namely recombinant protein, DNA, and overexpressing cells, we identified a chimeric anti-OX40 antibody (mAb035-hIgG1 from DNA immunization) that shows excellent binding specificity, and slightly stronger activation of human memory CD4+ T cells and similar potent antitumor activity compared with BMS 986178, an anti-OX40 antibody currently being evaluated for the treatment of solid tumors. This paper further systematically investigates the antigen-specific immune response, the number of binders, epitope bins, and functional activities of antibodies among different immunization strategies. Interestingly, we found that different immunization strategies affect the biological activity of monoclonal antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Inmunización/métodos , Receptores OX40/inmunología , Proteínas Recombinantes de Fusión/farmacología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/metabolismo , Bioensayo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Células CHO , Cricetulus , Femenino , Adyuvante de Freund/administración & dosificación , Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Hibridomas/química , Hibridomas/inmunología , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Fragmentos Fc de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fc de Inmunoglobulinas/farmacología , Células Jurkat , Luciferasas/genética , Luciferasas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/inmunología , Receptores OX40/antagonistas & inhibidores , Receptores OX40/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación
14.
Clin Cancer Res ; 27(15): 4435-4448, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34011561

RESUMEN

PURPOSE: Ovarian cancer represents a major clinical hurdle for immune checkpoint blockade (ICB), with reported low patient response rates. We found that the immune checkpoint ligand PD-L2 is robustly expressed in patient samples of ovarian cancers and other malignancies exhibiting suboptimal response to ICB but not in cancers that are ICB sensitive. Therefore, we hypothesize that PD-L2 can facilitate immune escape from ICB through incomplete blockade of the PD-1 signaling pathway. EXPERIMENTAL DESIGN: We engineered a soluble form of the PD-1 receptor (sPD-1) capable of binding and neutralizing both PD-L2 and PD-L1 with ×200 and ×10,000 folds improvement in binding affinity over wild-type PD-1 leading to superior inhibition of ligand-mediated PD-1 activities. RESULTS: Both in vitro and in vivo analyses performed in this study demonstrated that the high-affinity sPD-1 molecule is superior at blocking both PD-L1- and PD-L2-mediated immune evasion and reducing tumor growth in immune-competent murine models of ovarian cancer. CONCLUSIONS: The data presented in this study provide justification for using a dual targeting, high-affinity sPD-1 receptor as an alternative to PD-1 or PD-L1 therapeutic antibodies for achieving superior therapeutic efficacy in cancers expressing both PD-L2 and PD-L1.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Proteína 2 Ligando de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones
15.
J Cell Biol ; 161(5): 861-74, 2003 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-12796475

RESUMEN

Nuclear factor of activated T cell (NFAT) is a ubiquitous regulator involved in multiple biological processes. Here, we demonstrate that NFAT is temporally required in the developing atrial myocardium between embryonic day 14 and P0 (birth). Inhibition of NFAT activity by conditional expression of dominant-negative NFAT causes thinning of the atrial myocardium. The thin myocardium exhibits severe sarcomere disorganization and reduced expression of cardiac troponin-I (cTnI) and cardiac troponin-T (cTnT). Promoter analysis indicates that NFAT binds to and regulates transcription of the cTnI and the cTnT genes. Thus, regulation of cytoskeletal protein gene expression by NFAT may be important for the structural architecture of the developing atrial myocardium.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Atrios Cardíacos/anomalías , Atrios Cardíacos/crecimiento & desarrollo , Miocardio/metabolismo , Proteínas Nucleares , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Núcleo Celular/genética , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/genética , Feto , Genes Reguladores/genética , Atrios Cardíacos/metabolismo , Ratones , Ratones Transgénicos , Microscopía Electrónica , Mutación/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Factores de Transcripción NFATC , Regiones Promotoras Genéticas/genética , Sarcómeros/metabolismo , Sarcómeros/patología , Sarcómeros/ultraestructura , Factores de Transcripción/genética , Troponina I/biosíntesis , Troponina I/genética , Troponina T/biosíntesis , Troponina T/genética
16.
Mol Cell Biol ; 26(20): 7372-87, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16908540

RESUMEN

Compromised immunoregulation contributes to obesity and complications in metabolic pathogenesis. Here, we demonstrate that the nuclear factor of activated T cell (NFAT) group of transcription factors contributes to glucose and insulin homeostasis. Expression of two members of the NFAT family (NFATc2 and NFATc4) is induced upon adipogenesis and in obese mice. Mice with the Nfatc2-/- Nfatc4-/- compound disruption exhibit defects in fat accumulation and are lean. Nfatc2-/- Nfatc4-/- mice are also protected from diet-induced obesity. Ablation of NFATc2 and NFATc4 increases insulin sensitivity, in part, by sustained activation of the insulin signaling pathway. Nfatc2-/- Nfatc4-/- mice also exhibit an altered adipokine profile, with reduced resistin and leptin levels. Mechanistically, NFAT is recruited to the transcription loci and regulates resistin gene expression upon insulin stimulation. Together, these results establish a role for NFAT in glucose/insulin homeostasis and expand the repertoire of NFAT function to metabolic pathogenesis and adipokine gene transcription.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteínas Quinasas Activadas por AMP , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Animales , Diferenciación Celular , Línea Celular , Chlorocebus aethiops , Grasas de la Dieta/farmacología , Expresión Génica , Humanos , Ratones , Ratones Noqueados , Complejos Multienzimáticos/metabolismo , Factores de Transcripción NFATC/deficiencia , Factores de Transcripción NFATC/genética , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Resistina/metabolismo , Sensibilidad y Especificidad , Transducción de Señal
17.
Sci Rep ; 9(1): 17830, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780710

RESUMEN

Programmed cell death 1 (PD-1) monoclonal antibodies have been approved by regulatory agencies for the treatment of various types of cancer, and the mechanism involves the restoration of T cell functions. We report herein the X-ray crystal structure of a fully human monoclonal antibody mAb059c fragment antigen-binding (Fab) in complex with the PD-1 extracellular domain (ECD) at a resolution of 1.70 Å. Structural analysis indicates 1) an epitope, comprising fragments from the C'D, BC and FG loops of PD-1, contributes to mAb059c interaction, 2) an unique conformation of the C'D loop and a different orientation of R86 enabling the capture of PD-1 by the antibody complementarity determining region (CDR) and the formation of one salt-bridge contact - ASP101(HCDR3):ARG86(PD-1), and 3) the contact of FG with light chain (LC) CDR3 is maintained by a second salt-bridge and two backbone hydrogen bonds. Interface analysis reveals that N-glycosylation sites 49, 74 and 116 on PD-1 do not contact mAb059c; while N58 in the BC loop is recognized by mAb059c heavy chain CDR1 and CDR2. Mutation of N58 attenuated mAb059c binding to PD-1. These findings and the novel anti-PD-1 antibody will facilitate better understanding of the mechanisms of the molecular recognition of PD-1 receptor by anti-PD-1 mAb and, thereby, enable the development of new therapeutics with an expanded spectrum of efficacy for unmet medical needs.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias del Colon/terapia , Receptor de Muerte Celular Programada 1/química , Animales , Afinidad de Anticuerpos , Complejo Antígeno-Anticuerpo/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/química , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Enlace de Hidrógeno , Fragmentos Fab de Inmunoglobulinas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/genética , Unión Proteica , Dominios Proteicos , Transfección
18.
Mol Cell Biol ; 25(3): 907-20, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15657420

RESUMEN

Integration of protein kinases into transcription activation complexes influences the magnitude of gene expression. The nuclear factor of activated T cells (NFAT) group of proteins are critical transcription factors that direct gene expression in immune and nonimmune cells. A balance of phosphotransferase activity is necessary for optimal NFAT activation. Activation of NFAT requires dephosphorylation by the calcium-mediated calcineurin phosphatase to promote NFAT nuclear accumulation, and the Ras-activated extracellular signal-regulated kinase (ERK) mitogen-activated protein (MAP) kinase, which targets NFAT partners, to potentiate transcription. Whether protein kinases operate on NFAT and contribute positively to transcription activation is not clear. Here, we coupled DNA affinity isolation with in-gel kinase assays to avidly pull down the activated NFAT and identify its associated protein kinases. We demonstrate that p90 ribosomal S6 kinase (RSK) is recruited to the NFAT-DNA transcription complex upon activation. The formation of RSK-NFATc4-DNA transcription complex is also apparent upon adipogenesis. Bound RSK phosphorylates Ser(676) and potentiates NFATc4 DNA binding by escalating NFAT-DNA association. Ser(676) is also targeted by the ERK MAP kinase, which interacts with NFAT at a distinct region than RSK. Thus, integration of the ERK/RSK signaling pathway provides a mechanism to modulate NFATc4 transcription activity.


Asunto(s)
Calcineurina/metabolismo , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Factores de Transcripción/metabolismo , Células 3T3-L1 , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Activación Enzimática/fisiología , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Transcripción NFATC , Fosforilación , Unión Proteica , Transducción de Señal/fisiología
19.
Mol Cell Biol ; 22(11): 3892-904, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11997522

RESUMEN

Nuclear factor of activated T cells (NFAT) is implicated in multiple biological processes, including cytokine gene expression, cardiac hypertrophy, and adipocyte differentiation. A conserved NFAT homology domain is identified in all NFAT members. Dephosphorylation of the NFAT homology region is critical for NFAT nuclear translocation and transcriptional activation. Here we demonstrate that NFATc4 is phosphorylated by p38 mitogen-activated protein (MAP) kinase but not by JNK. The p38 MAP kinase phosphorylates multiple residues, including Ser(168) and Ser(170), in the NFAT homology domain of NFATc4. Replacement of Ser(168,170) with Ala promotes nuclear localization of NFATc4 and increases NFAT-mediated transcription activity. Stable expression of Ala(168,170) NFATc4, but not of wild-type NFATc4, in NIH 3T3 cells promotes adipocyte formation under differentiation conditions. Molecular analysis indicates that peroxisome proliferator-activated receptor gamma 2 (PPAR gamma 2) is a target of NFAT. Two distinct NFAT binding elements are located in the PPAR gamma 2 gene promoter. Stable expression of Ala(168,170) NFATc4, but not of wild-type NFATc4, increases the expression of PPAR gamma, which contributes in part to increased adipocyte formation. Thus, NFAT regulates PPAR gamma gene expression and has a direct role in adipocyte differentiation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares , Factores de Transcripción/metabolismo , Células 3T3 , Adipocitos/citología , Adipocitos/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Células COS , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Secuencia Conservada , Cricetinae , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Factores de Transcripción NFATC , Fosforilación , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/genética , Homología de Secuencia de Aminoácido , Serina/química , Factores de Transcripción/química , Factores de Transcripción/genética , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Diabetes ; 51(10): 3049-54, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12351446

RESUMEN

Diabetes is a major risk factor for premature atherosclerosis, and oxidative stress appears to be an important mechanism. Previously, we showed that diabetic monocytes produce increased superoxide anion (O(2)(-)), and alpha-tocopherol (AT) supplementation decreases this. The aim of this study was to elucidate the mechanism(s) of O(2)(-) release and inhibition by AT under hyperglycemic (HG) conditions in monocytes. O(2)(-) release, protein kinase C (PKC) activity, and translocation of PKC-alpha and -betaII and p47phox were increased in THP-1 cells (human monocytic cell line) under HG (15 mmol/l glucose) conditions, whereas AT supplementation inhibited these changes. AT, NADPH oxidase inhibitors (apocynin and diphenyleneiodonium chloride [DPI]), and an inhibitor to PKC-alpha and other isoforms (2,2',3,3',4,4'-hexahydroxy-1,1'-biphenyl-6,6'-dimethanol dimethyl ether [HBDDE]) but not PKC-beta II (LY379196) decreased O(2)(-) release and p47phox translocation. Antisense oligodeoxynucleotides to PKC-alpha and p47phox but not to PKC-betaII inhibited HG-induced O(2)(-) release and p47phox translocation in THP-1 cells. Under HG conditions, reactive oxygen species release from monocytes was not inhibited by agents affecting mitochondrial metabolism but was inhibited in human endothelial cells. We conclude that under HG conditions, monocytic O(2)(-) release is dependent on NADPH oxidase activity but not the mitochondrial respiratory chain; HG-induced O(2)(-) release is triggered by PKC-alpha, and AT inhibits O(2)(-) release via inhibition of PKC-alpha.


Asunto(s)
Antioxidantes/farmacología , Ácido Elágico/análogos & derivados , Hiperglucemia/metabolismo , Isoenzimas/metabolismo , Monocitos/enzimología , Proteína Quinasa C/metabolismo , Superóxidos/metabolismo , alfa-Tocoferol/farmacología , Acetofenonas/farmacología , Compuestos de Bifenilo/farmacología , Línea Celular , Ácido Elágico/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Mesilatos/farmacología , Monocitos/citología , NADPH Oxidasas , Oligodesoxirribonucleótidos Antisentido/farmacología , Compuestos Onio/farmacología , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C beta , Proteína Quinasa C-alfa , Pirroles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA