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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioconjug Chem ; 35(6): 855-866, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38789102

RESUMEN

Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.


Asunto(s)
Receptores de IgG , Animales , Ratones , Receptores de IgG/inmunología , Humanos , Polietilenglicoles/química , Citotoxicidad Celular Dependiente de Anticuerpos , Fagocitosis/efectos de los fármacos
2.
Mol Cancer Ther ; 23(1): 68-83, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37775098

RESUMEN

Brentuximab vedotin, a CD30-directed antibody-drug conjugate (ADC), is approved for clinical use in multiple CD30-expressing lymphomas. The cytotoxic payload component of brentuximab vedotin is monomethyl auristatin E (MMAE), a highly potent microtubule-disrupting agent. Preclinical results provided here demonstrate that treatment of cancer cells with brentuximab vedotin or free MMAE leads to a catastrophic disruption of the microtubule network eliciting a robust endoplasmic reticulum (ER) stress response that culminates in the induction of the classic hallmarks of immunogenic cell death (ICD). In accordance with the induction of ICD, brentuximab vedotin-killed lymphoma cells drove innate immune cell activation in vitro and in vivo. In the "gold-standard" test of ICD, vaccination of mice with brentuximab vedotin or free MMAE-killed tumor cells protected animals from tumor rechallenge; in addition, T cells transferred from previously vaccinated animals slowed tumor growth in immunodeficient mice. Immunity acquired from killed tumor cell vaccination was further amplified by the addition of PD-1 blockade. In a humanized model of CD30+ B-cell tumors, treatment with brentuximab vedotin drove the expansion and recruitment of autologous Epstein-Barr virus-reactive CD8+ T cells potentiating the activity of anti-PD-1 therapy. Together, these data support the ability of brentuximab vedotin and MMAE to drive ICD in tumor cells resulting in the activation of antigen-presenting cells and augmented T-cell immunity. These data provide a strong rationale for the clinical combination of brentuximab vedotin and other MMAE-based ADCs with checkpoint inhibitors.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Inmunoconjugados , Animales , Ratones , Brentuximab Vedotina , Muerte Celular Inmunogénica , Antígeno Ki-1 , Herpesvirus Humano 4/metabolismo , Inmunoconjugados/uso terapéutico , Microtúbulos/metabolismo
3.
J Neurosci ; 32(8): 2696-702, 2012 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-22357853

RESUMEN

Several anti-amyloid ß (Aß) antibodies are under evaluation for the treatment of Alzheimer's disease (AD). Clinical studies using the N-terminal-directed anti-Aß antibody bapineuzumab have demonstrated reduced brain PET-Pittsburg-B signals, suggesting the reduction of Aß plaques, and reduced levels of total and phosphorylated tau protein in the CSF of treated AD patients. Preclinical studies using 3D6 (the murine form of bapineuzumab) have demonstrated resolution of Aß plaque and vascular burdens, neuritic dystrophy, and preservation of synaptic density in the transgenic APP mouse models. In contrast, few studies have evaluated the direct interaction of this antibody with synaptotoxic soluble Aß species. In the current report, we demonstrated that 3D6 binds to soluble, synaptotoxic assemblies of Aß(1-42) and prevents multiple downstream functional consequences in rat hippocampal neurons including changes in glutamate AMPA receptor trafficking, AD-type tau phosphorylation, and loss of dendritic spines. In vivo, we further demonstrated that 3D6 prevents synaptic loss and acutely reverses the behavioral deficit in the contextual fear conditioning task in transgenic mouse models of AD, two endpoints thought to be linked to synaptotoxic soluble Aß moieties. Importantly C-terminal anti-Aß antibodies were ineffective on these endpoints. These results, taken with prior studies, suggest that N-terminal anti-Aß antibodies effectively interact with both soluble and insoluble forms of Aß and therefore appear particularly well suited for testing the Aß hypothesis of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/inmunología , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Epítopos/inmunología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Anticuerpos Neutralizantes , Síntomas Conductuales/tratamiento farmacológico , Síntomas Conductuales/etiología , Síntomas Conductuales/inmunología , Biotina/metabolismo , Células Cultivadas , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Espinas Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epítopos/metabolismo , Miedo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/citología , Humanos , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/inmunología , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/inmunología , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/inmunología , Neuropéptidos/metabolismo , Fragmentos de Péptidos/inmunología , Fosforilación , Unión Proteica/inmunología , Estructura Secundaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores AMPA/metabolismo , Solubilidad , Proteína 1 de Transporte Vesicular de Glutamato/inmunología , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
4.
Front Immunol ; 14: 1280986, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022590

RESUMEN

TIGIT is an immune checkpoint receptor expressed on activated and memory T cells, immunosuppressive T regulatory cells, and natural killer (NK) cells. TIGIT has emerged as an attractive target for antitumor therapies, due to its proposed immunosuppressive effects on lymphocyte function and T cell activation. We generated an anti-TIGIT monoclonal antibody (mAb) that binds with high affinity to human, non-human primate, and murine TIGIT and through multiple experimental methodologies demonstrated that checkpoint blockade alone is insufficient for antitumor activity. Generating anti-TIGIT mAbs with various Fc backbones we show that muting the Fc-Fcγ receptor (FcγR) interaction failed to drive antitumor activity, while mAbs with Fc functional backbones demonstrate substantial antitumor activity, mediated through activation of antigen-presenting cells (APCs), T cell priming, and NK-mediated depletion of suppressive Tregs and exhausted T cells. Further, nonfucosylation of the Fc backbone resulted in enhanced immune responses and antitumor activity relative to the intact IgG1 backbone. The improved activity correlated with the biased FcγR interaction profile of the nonfucosylated anti-TIGIT mAb, which supports that FcγRIIIa binding with decreased FcγRIIb binding favorably activates APCs and enhances tumor-specific CD8+ T cell responses. The anti-TIGIT mAbs with intact FcγR interacting backbones also demonstrated synergistic enhancement of other standard antitumor treatments, including anti-PD-1 treatment and a model monomethyl auristatin E antibody-drug conjugate. These findings highlight the importance of the anti-TIGIT mAb's Fc backbone to its antitumor activity and the extent to which this activity can be enhanced through nonfucosylation of the backbone.


Asunto(s)
Neoplasias , Receptores de IgG , Ratones , Animales , Receptores Inmunológicos/metabolismo , Anticuerpos Monoclonales/farmacología , Inmunidad Innata
5.
J Immunother Cancer ; 11(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37793853

RESUMEN

BACKGROUND: SGN-B7H4V is a novel investigational vedotin antibody-drug conjugate (ADC) comprising a B7-H4-directed human monoclonal antibody conjugated to the cytotoxic payload monomethyl auristatin E (MMAE) via a protease-cleavable maleimidocaproyl valine citrulline (mc-vc) linker. This vedotin linker-payload system has been clinically validated in multiple Food and Drug Administration approved agents including brentuximab vedotin, enfortumab vedotin, and tisotumab vedotin. B7-H4 is an immune checkpoint ligand with elevated expression on a variety of solid tumors, including breast, ovarian, and endometrial tumors, and limited normal tissue expression. SGN-B7H4V is designed to induce direct cytotoxicity against target cells by binding to B7-H4 on the surface of target cells and releasing the cytotoxic payload MMAE upon internalization of the B7-H4/ADC complex. METHODS: B7-H4 expression was characterized by immunohistochemistry across multiple solid tumor types. The ability of SGN-B7H4V to kill B7-H4-expressing tumor cells in vitro and in vivo in a variety of xenograft tumor models was also evaluated. Finally, the antitumor activity of SGN-B7H4V as monotherapy and in combination with an anti-programmed cell death-1 (PD-1) agent was evaluated using an immunocompetent murine B7-H4-expressing Renca tumor model. RESULTS: Immunohistochemistry confirmed B7-H4 expression across multiple solid tumors, with the highest prevalence in breast, endometrial, and ovarian tumors. In vitro, SGN-B7H4V killed B7-H4-expressing tumor cells by MMAE-mediated direct cytotoxicity and antibody-mediated effector functions including antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. In vivo, SGN-B7H4V demonstrated strong antitumor activity in multiple xenograft models of breast and ovarian cancer, including xenograft tumors with heterogeneous B7-H4 expression, consistent with the ability of vedotin ADCs to elicit a bystander effect. In an immunocompetent murine B7-H4-expressing tumor model, SGN-B7H4V drove robust antitumor activity as a monotherapy that was enhanced when combined with an anti-PD-1 agent. CONCLUSION: The immune checkpoint ligand B7-H4 is a promising molecular target expressed by multiple solid tumors. SGN-B7H4V demonstrates robust antitumor activity in preclinical models through multiple potential mechanisms. Altogether, these preclinical data support the evaluation of SGN-B7H4V as a monotherapy in the ongoing phase 1 study of SGN-B7H4V in advanced solid tumors (NCT05194072) and potential future clinical combinations with immunotherapies.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Animales , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Inmunoconjugados/química , Inmunohistoquímica , Ligandos
6.
J Cell Biol ; 174(4): 547-56, 2006 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-16908670

RESUMEN

The mammalian ATP-binding cassette transporters A1 and A7 (ABCA1 and -A7) show sequence similarity to CED-7, a Caenorhabditis elegans gene that mediates the clearance of apoptotic cells. Using RNA interference or gene targeting, we show that knock down of macrophage ABCA7 but not -A1 results in defective engulfment of apoptotic cells. In response to apoptotic cells, ABCA7 moves to the macrophage cell surface and colocalizes with the low-density lipoprotein receptor-related protein 1 (LRP1) in phagocytic cups. The cell surface localization of ABCA7 and LRP1 is defective in ABCA7-deficient cells. C1q is an opsonin of apoptotic cells that acts via phagocyte LRP1 to induce extracellular signal-regulated kinase (ERK) signaling. We show that ERK signaling is required for phagocytosis of apoptotic cells and that ERK phosphorylation in response to apoptotic cells or C1q is defective in ABCA7-deficient cells. These studies reveal a major role of ABCA7 and not -A1 in the clearance of apoptotic cells and therefore suggest that ABCA7 is an authentic orthologue of CED-7.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Apoptosis/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Macrófagos/metabolismo , Fagocitosis/fisiología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Complemento C1q/metabolismo , Regulación hacia Abajo/fisiología , Femenino , Marcación de Gen , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba/fisiología
7.
Mol Cancer Ther ; 19(5): 1102-1109, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32165557

RESUMEN

2-fluorofucose (2FF) inhibits protein and cellular fucosylation. Afucosylation of IgG antibodies enhances antibody-dependent cell-mediated cytotoxicity by modulating antibody affinity for FcγRIIIa, which can impact secondary T-cell activation. Immune responses toward most common solid tumors are dominated by a humoral immune response rather than the presence of tumor-infiltrating cytotoxic T cells. IgG antibodies directed against numerous tumor-associated proteins are found in the sera of both patients with breast cancer and transgenic mice bearing mammary cancer. We questioned whether 2FF would have antitumor activity in two genetically distinct transgenic models; TgMMTV-neu (luminal B) and C3(1)-Tag (basal) mammary cancer. 2FF treatment significantly improved overall survival. The TgMMTV-neu doubled survival time compared with controls [P < 0.0001; HR, 7.04; 95% confidence interval (CI), 3.31-15.0], and survival was significantly improved in C3(1)-Tag (P = 0.0013; HR, 3.36; 95% CI, 1.58-7.14). 2FF treated mice, not controls, developed delayed-type hypersensitivity and T-cell responses specific for syngeneic tumor lysates (P < 0.0001). Serum IgG from 2FF-treated mice enhanced tumor lysis more efficiently than control sera (P = 0.004). Administration of 2FF for prophylaxis, at two different doses, significantly delayed tumor onset in both TgMMTV-neu; 20 mmol/L (P = 0.0004; HR, 3.55; 95% CI, 1.60-7.88) and 50 mmol/L (P = 0.0002; HR: 3.89; 95% CI, 1.71-8.86) and C3(1)-Tag; 20 mmol/L (P = 0.0020; HR, 2.51; 95% CI, 1.22-5.18), and 50 mmol/L (P = 0.0012; HR, 3.36; 95% CI, 1.57-7.18). Mammary cancer was prevented in 33% of TgMMTV-neu and 26% of C3(1)-Tag. 2FF has potent antitumor effects in mammary cancer models. The agent shows preclinical efficacy for both cancer treatment and prevention.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Fucosa/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Animales , Apoptosis , Proliferación Celular , Femenino , Fucosa/administración & dosificación , Glicosilación , Humanos , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Células Tumorales Cultivadas
8.
Am J Respir Crit Care Med ; 178(2): 158-67, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18420961

RESUMEN

RATIONALE: Efficient removal of apoptotic cells is essential for the resolution of acute pulmonary inflammation. Alveolar macrophages ingest apoptotic cells less avidly than other professional phagocytes at rest but overcome this defect during acute inflammation. Surfactant protein (SP)-A and SP-D are potent modulators of macrophage function and may suppress clearance of apoptotic cells through activation of the transmembrane receptor signal inhibitory regulatory protein alpha (SIRP alpha). OBJECTIVES: To investigate whether binding of SP-A and SP-D to SIRP alpha on alveolar macrophages suppresses apoptotic cell clearance. METHODS: Phagocytosis of apoptotic cells was assessed using macrophages pretreated with SP-A, SP-D, or the collectin-like molecule C1q. Binding of SP-A and SP-D to SIRP alpha was confirmed in vitro using blocking antibodies and fibroblasts transfected with active and mutant SIRP alpha. The effects of downstream molecules SHP-1 and RhoA on phagocytosis were studied using SHP-1-deficient mice, sodium stibogluconate, and a Rho kinase inhibitor. Lipopolysaccharide was given to chimeric mice to study the effects of SP-A and SP-D binding on inflammatory macrophages. MEASUREMENTS AND MAIN RESULTS: Preincubation of macrophages with SP-A or SP-D suppressed apoptotic cell clearance. Surfactant suppression of macrophage phagocytosis was reversed by blocking SIRP alpha and inhibiting downstream molecules SHP-1 and RhoA. Macrophages from inflamed lungs ingested apoptotic cells more efficiently than resting alveolar macrophages. Recruited mononuclear phagocytes with low levels of SP-A and SP-D mediated this effect. CONCLUSIONS: SP-A and SP-D tonically inhibit alveolar macrophage phagocytosis by binding SIRP alpha. During acute pulmonary inflammation, defects in apoptotic cell clearance are overcome by recruited mononuclear phagocytes.


Asunto(s)
Antígenos de Diferenciación/inmunología , Inflamación/fisiopatología , Macrófagos Alveolares/inmunología , Fagocitosis/inmunología , Proteína A Asociada a Surfactante Pulmonar/inmunología , Proteína D Asociada a Surfactante Pulmonar/inmunología , Receptores Inmunológicos/inmunología , Animales , Apoptosis/inmunología , Unión Competitiva , Células Cultivadas , Humanos , Inflamación/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
9.
Sci Rep ; 9(1): 16947, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31740740

RESUMEN

The over-expression and aggregation of α-synuclein (αSyn) are linked to the onset and pathology of Parkinson's disease. Native monomeric αSyn exists in an intrinsically disordered ensemble of interconverting conformations, which has made its therapeutic targeting by small molecules highly challenging. Nonetheless, here we successfully target the monomeric structural ensemble of αSyn and thereby identify novel drug-like small molecules that impact multiple pathogenic processes. Using a surface plasmon resonance high-throughput screen, in which monomeric αSyn is incubated with microchips arrayed with tethered compounds, we identified novel αSyn interacting drug-like compounds. Because these small molecules could impact a variety of αSyn forms present in the ensemble, we tested representative hits for impact on multiple αSyn malfunctions in vitro and in cells including aggregation and perturbation of vesicular dynamics. We thereby identified a compound that inhibits αSyn misfolding and is neuroprotective, multiple compounds that restore phagocytosis impaired by αSyn overexpression, and a compound blocking cellular transmission of αSyn. Our studies demonstrate that drug-like small molecules that interact with native αSyn can impact a variety of its pathological processes. Thus, targeting the intrinsically disordered ensemble of αSyn offers a unique approach to the development of small molecule research tools and therapeutics for Parkinson's disease.


Asunto(s)
Bibliotecas de Moléculas Pequeñas/farmacología , alfa-Sinucleína/metabolismo , Amiloide/antagonistas & inhibidores , Amiloide/metabolismo , Línea Celular , Transferencia Resonante de Energía de Fluorescencia , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Proteínas Intrínsecamente Desordenadas/metabolismo , Fagocitosis/efectos de los fármacos , Pliegue de Proteína , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/toxicidad , Resonancia por Plasmón de Superficie , alfa-Sinucleína/química , alfa-Sinucleína/efectos de los fármacos
10.
J Leukoc Biol ; 79(5): 896-903, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16641135

RESUMEN

The process of apoptosis includes critically important changes on the cell surface that lead to its recognition and removal. The recognition also generates a number of other local tissue responses including suppression of inflammation and immunity. It is surprising that the ligands generated on the apoptotic cell, which mediates these effects, have received relatively little attention. Some of these candidate molecules and possible mechanisms for their surface expression are addressed herein, with particular emphasis on phosphatidylserine and calreticulin. However, exposure of such ligands is exclusive to apoptosis and may, in fact, occur on viable cells. To partially explain the lack of response to such potential stimuli, the presence on viable cells of "don't eat me" signals, in this case, CD47 is suggested to prevent such unwarranted actions. Loss or inactivation of the don't eat me CD47 effects accompanies apoptosis and now allow the cells to be recognized and cleared.


Asunto(s)
Antígenos de Superficie/inmunología , Apoptosis/inmunología , Antígeno CD47/inmunología , Comunicación Celular/inmunología , Transducción de Señal/inmunología , Animales , Antígenos de Superficie/química , Antígeno CD47/química , Calreticulina/inmunología , Colectinas/metabolismo , Humanos , Ligandos , Fosfatidilserinas/inmunología
11.
Circ Res ; 92(10): 1162-9, 2003 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12714563

RESUMEN

PPARgamma is a member of a family of nuclear receptors/ligand-dependent transcription factors, which bind to hormone response elements on target gene promoters. An antiproliferative and proapoptotic action profile of PPARgamma has been described and PPARgamma may function as a tumor suppressor gene, but little is known about the role of PPARgamma in vascular remodeling. One group of human diseases that shows impressive vascular remodeling exclusively in the lungs is the group of severe pulmonary hypertensive disorders, which is characterized by complex, endothelial cell-proliferative lesions of lung precapillary arterioles composed of clusters of phenotypically altered endothelial cells that occlude the vessel lumen and contribute to the elevation of the pulmonary arterial pressure and reduce local lung tissue blood flow. In the present study, we report the ubiquitous PPARgamma expression in normal lungs, and in contrast, a reduced lung tissue PPARgamma gene and protein expression in the lungs from patients with severe PH and loss of PPARgamma expression in their complex vascular lesions. We show that fluid shear stress reduces PPARgamma expression in ECV304 endothelial cells, that ECV304 cells that stably express dominant-negative PPARgamma (DN-PPARgamma ECV304) form sprouts when placed in matrigel and that DN-PPARgamma ECV304 cells, after tail vein injection in nude mice, form lumen-obliterating lung vascular lesions. We conclude that fluid shear stress decreases the expression of PPARgamma in endothelial cells and that loss of PPARgamma expression characterizes an abnormal, proliferating, apoptosis-resistant endothelial cell phenotype.


Asunto(s)
Endotelio Vascular/metabolismo , Hipertensión Pulmonar/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , División Celular , Línea Celular , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/trasplante , Femenino , Expresión Génica , Genes Dominantes , Humanos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/patología , Inmunohistoquímica , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Pulmón/patología , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/genética , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/patología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética , Estrés Mecánico , Factores de Transcripción/genética , Transfección
12.
PLoS One ; 9(8): e102909, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25170892

RESUMEN

Detailed analysis of disease-affected tissue provides insight into molecular mechanisms contributing to pathogenesis. Substantia nigra, striatum, and cortex are functionally connected with increasing degrees of alpha-synuclein pathology in Parkinson's disease. We undertook functional and causal pathway analysis of gene expression and proteomic alterations in these three regions, and the data revealed pathways that correlated with disease progression. In addition, microarray and RNAseq experiments revealed previously unidentified causal changes related to oligodendrocyte function and synaptic vesicle release, and these and other changes were reflected across all brain regions. Importantly, subsets of these changes were replicated in Parkinson's disease blood; suggesting peripheral tissue may provide important avenues for understanding and measuring disease status and progression. Proteomic assessment revealed alterations in mitochondria and vesicular transport proteins that preceded gene expression changes indicating defects in translation and/or protein turnover. Our combined approach of proteomics, RNAseq and microarray analyses provides a comprehensive view of the molecular changes that accompany functional loss and alpha-synuclein pathology in Parkinson's disease, and may be instrumental to understand, diagnose and follow Parkinson's disease progression.


Asunto(s)
Encéfalo/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Animales , Encéfalo/metabolismo , Progresión de la Enfermedad , Regulación de la Expresión Génica , Humanos , Análisis por Micromatrices , Proteínas/análisis , Proteínas/genética , Proteínas/metabolismo , Proteómica , Análisis de Secuencia de ARN , Transducción de Señal , alfa-Sinucleína/análisis , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
13.
PLoS One ; 9(2): e87133, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24551051

RESUMEN

The misfolding of intrinsically disordered proteins such as α-synuclein, tau and the Aß peptide has been associated with many highly debilitating neurodegenerative syndromes including Parkinson's and Alzheimer's diseases. Therapeutic targeting of the monomeric state of such intrinsically disordered proteins by small molecules has, however, been a major challenge because of their heterogeneous conformational properties. We show here that a combination of computational and experimental techniques has led to the identification of a drug-like phenyl-sulfonamide compound (ELN484228), that targets α-synuclein, a key protein in Parkinson's disease. We found that this compound has substantial biological activity in cellular models of α-synuclein-mediated dysfunction, including rescue of α-synuclein-induced disruption of vesicle trafficking and dopaminergic neuronal loss and neurite retraction most likely by reducing the amount of α-synuclein targeted to sites of vesicle mobilization such as the synapse in neurons or the site of bead engulfment in microglial cells. These results indicate that targeting α-synuclein by small molecules represents a promising approach to the development of therapeutic treatments of Parkinson's disease and related conditions.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/antagonistas & inhibidores , Terapia Molecular Dirigida , Enfermedad de Parkinson/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , alfa-Sinucleína/antagonistas & inhibidores , Animales , Sitios de Unión , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Ratones , Modelos Biológicos , Modelos Moleculares , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Enfermedad de Parkinson/patología , Fagocitos/efectos de los fármacos , Fagocitos/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo
14.
PLoS One ; 8(8): e71634, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24058406

RESUMEN

Alpha-synuclein protein is strongly implicated in the pathogenesis Parkinson's disease. Increased expression of α-synuclein due to genetic multiplication or point mutations leads to early onset disease. While α-synuclein is known to modulate membrane vesicle dynamics, it is not clear if this activity is involved in the pathogenic process or if measurable physiological effects of α-synuclein over-expression or mutation exist in vivo. Macrophages and microglia isolated from BAC α-synuclein transgenic mice, which overexpress α-synuclein under regulation of its own promoter, express α-synuclein and exhibit impaired cytokine release and phagocytosis. These processes were affected in vivo as well, both in peritoneal macrophages and microglia in the CNS. Extending these findings to humans, we found similar results with monocytes and fibroblasts isolated from idiopathic or familial Parkinson's disease patients compared to age-matched controls. In summary, this paper provides 1) a new animal model to measure α-synuclein dysfunction; 2) a cellular system to measure synchronized mobilization of α-synuclein and its functional interactions; 3) observations regarding a potential role for innate immune cell function in the development and progression of Parkinson's disease and other human synucleinopathies; 4) putative peripheral biomarkers to study and track these processes in human subjects. While altered neuronal function is a primary issue in PD, the widespread consequence of abnormal α-synuclein expression in other cell types, including immune cells, could play an important role in the neurodegenerative progression of PD and other synucleinopathies. Moreover, increased α-synuclein and altered phagocytosis may provide a useful biomarker for human PD.


Asunto(s)
Inmunidad Innata , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/inmunología , alfa-Sinucleína/inmunología , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Citocinas/inmunología , Femenino , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Transgénicos , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Fagocitosis , Regulación hacia Arriba , alfa-Sinucleína/genética
15.
J Biol Chem ; 281(50): 38376-84, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17056601

RESUMEN

Apoptotic cells are rapidly engulfed by adjacent tissue cells or macrophages before they can release pro-inflammatory/proimmunogenic intracellular contents. In addition, recognition of the apoptotic cells is actively anti-inflammatory and anti-immunogenic with generation of anti-inflammatory mediators such as transforming growth factor-beta (TGF-beta) and anti-inflammatory eicosanoids. Here, we have investigated the role played by the induction of TGF-beta in the coordinate expression of anti-inflammatory eicosanoids or peroxisome proliferator-activated receptor-gamma and in the suppression of pro-inflammatory lipid mediators and nitric oxide (NO). By use of a dominant negative TGFbetaII receptor, TGF-beta signaling was blocked, and its participation in the consequences of apoptotic cell stimulation was determined. The induction of TGF-beta itself could be attributed to exposed phosphatidylserine on the apoptotic cells, which therefore appears to drive the balanced inflammatory mediator responses. Arachidonic acid release, COX-2, and prostaglandin synthase expression were shown to be significantly dependent on the TGF-beta production. On the other hand, a requirement for TGF-beta was also shown in the inhibition of thromboxane synthase and thromboxanes, of 5-lipoxygenase and sulfidopeptide leukotrienes, as well as of inducible nitric-oxide synthase and NO. TGF-beta-dependent induction of arginase was also found and would further limit the NO generation. Finally, apoptotic cells stimulated production of 15-lipoxygenase and 15-hydroxyeicosatetraenoic acid, a potentially anti-inflammatory pathway acting through peroxisome proliferator-activated receptor-gamma, and lipoxin A(4) production, which were also up-regulated by a TGF-beta-dependent pathway in this system. These results strongly suggest that the apoptotic cell inhibition of pro-inflammatory mediator production is pleiotropic and significantly dependent on the stimulation of TGF-beta production.


Asunto(s)
Apoptosis , Eicosanoides/biosíntesis , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/metabolismo , Óxido Nítrico/biosíntesis , Factor de Crecimiento Transformador beta/fisiología , Animales , Ácido Araquidónico/metabolismo , Ciclooxigenasa 2/metabolismo , Humanos , Células Jurkat , Ratones , Ratones Endogámicos BALB C , Transducción de Señal
16.
Cell ; 123(2): 321-34, 2005 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-16239148

RESUMEN

Apoptotic-cell removal is critical for development, tissue homeostasis, and resolution of inflammation. Although many candidate systems exist, only phosphatidylserine has been identified as a general recognition ligand on apoptotic cells. We demonstrate here that calreticulin acts as a second general recognition ligand by binding and activating LDL-receptor-related protein (LRP) on the engulfing cell. Since surface calreticulin is also found on viable cells, a mechanism preventing inadvertent uptake was sought. Disruption of interactions between CD47 (integrin-associated protein) on the target cell and SIRPalpha (SHPS-1), a heavily glycosylated transmembrane protein on the engulfing cell, permitted uptake of viable cells in a calreticulin/LRP-dependent manner. On apoptotic cells, CD47 was altered and/or lost and no longer activated SIRPalpha. These changes on the apoptotic cell create an environment where "don't eat me" signals are rendered inactive and "eat me" signals, including calreticulin and phosphatidylserine, congregate together and signal for removal.


Asunto(s)
Apoptosis/inmunología , Calreticulina/metabolismo , Fagocitos/inmunología , Receptores Inmunológicos/metabolismo , Activación Transcripcional , Animales , Anticuerpos Monoclonales/metabolismo , Apoptosis/genética , Antígeno CD47/metabolismo , Línea Celular , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/metabolismo , Heterocigoto , Humanos , Células Jurkat , Macrófagos/inmunología , Ratones , Ratones Noqueados , Microscopía por Video , Modelos Biológicos , Neutrófilos/citología , Neutrófilos/metabolismo , Fagocitosis/genética , Fagocitosis/inmunología , Receptores Inmunológicos/genética
17.
J Biol Chem ; 279(43): 44695-703, 2004 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-15292176

RESUMEN

Normal spontaneous apoptosis in neutrophils is enhanced by "stress" stimuli such as tumor necrosis factor-alpha, Fas ligand, and oxidants, and this effect is inhibited by anti-apoptotic stimuli including granulocyte-macrophage colony-stimulating factor, lipopolysaccharide, and formylmethionine-leucine-phenylalanine. In this report we demonstrate that anti-apoptotic stimuli protect neutrophils from stress-induced apoptosis via activation of the ERK/MAPK pathway. The protection occurs downstream of mitochondrial alterations assessed as a decrease in membrane potential concomitant with enhanced cytochrome c release. ERK activation was shown to inhibit apoptosis by maintaining levels of XIAP, which is normally decreased in the presence of the pro-apoptotic/stress stimuli. This report also demonstrates that potent intra- and extracellular oxidants inhibit the protective effect of ERK. Oxidant-dependent inhibition of ERK was because of activation of p38 MAPK and activation of the protein phosphatases PP1 and PP2A. Our data suggest that ERK suppresses stress-induced apoptosis downstream of mitochondrial alterations by maintaining XIAP levels and that oxidants block this effect through activation of p38 and protein phosphatases.


Asunto(s)
Apoptosis , Sistema de Señalización de MAP Quinasas , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oxidantes/metabolismo , Proteínas/metabolismo , Antioxidantes/metabolismo , Western Blotting , Células Cultivadas , Citocromos c/metabolismo , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Flavonoides/farmacología , Citometría de Flujo , Humanos , Lipopolisacáridos/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Neutrófilos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Factores de Tiempo , Rayos Ultravioleta , Proteína Inhibidora de la Apoptosis Ligada a X , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cell ; 115(1): 13-23, 2003 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-14531999

RESUMEN

Surfactant proteins A and D (SP-A and SP-D) are lung collectins composed of two regions, a globular head domain that binds PAMPs and a collagenous tail domain that initiates phagocytosis. We provide evidence that SP-A and SP-D act in a dual manner, to enhance or suppress inflammatory mediator production depending on binding orientation. SP-A and SP-D bind SIRPalpha through their globular heads to initiate a signaling pathway that blocks proinflammatory mediator production. In contrast, their collagenous tails stimulate proinflammatory mediator production through binding to calreticulin/CD91. Together a model is implied in which SP-A and SP-D help maintain a non/anti-inflammatory lung environment by stimulating SIRPalpha on resident cells through their globular heads. However, interaction of these heads with PAMPs on foreign organisms or damaged cells and presentation of the collagenous tails in an aggregated state to calreticulin/CD91, stimulates phagocytosis and proinflammatory responses.


Asunto(s)
Antígenos de Diferenciación , Calreticulina/metabolismo , Colectinas/metabolismo , Inflamación/metabolismo , Pulmón/metabolismo , Glicoproteínas de Membrana/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Receptores Inmunológicos , Animales , Calreticulina/inmunología , Células Cultivadas , Colectinas/química , Colectinas/inmunología , Complemento C1q/metabolismo , Citocinas/metabolismo , Activación Enzimática , Humanos , Péptidos y Proteínas de Señalización Intracelular , Pulmón/citología , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/inmunología , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteína A Asociada a Surfactante Pulmonar/química , Proteína A Asociada a Surfactante Pulmonar/inmunología , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/química , Proteína D Asociada a Surfactante Pulmonar/inmunología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
19.
J Biol Chem ; 279(20): 21085-95, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-14766748

RESUMEN

Although important for apoptosis, the mechanism of Bax regulation is poorly understood. This study demonstrates that phosphorylation of Ser(184) regulates Bax activity. The phosphorylation required phosphatidylinositol 3-kinase/Akt activation and appeared to be mediated by Akt itself. In the serine-phosphorylated form, Bax was detected in the cytoplasm, could not be immunoprecipitated with the activation-specific antibody 6A7, and promoted heterodimerization with Mcl-1, Bcl-x(L), and A1. Apoptotic neutrophils possessed reduced levels of serine-phosphorylated Bax correlating with an increase in activated Bax as well as an increase in the amount of Bax found translocated to the mitochondria. We suggest that Bax is regulated by phosphorylation of Ser(184) in an Akt-dependent manner and that phosphorylation inhibits Bax effects on the mitochondria by maintaining the protein in the cytoplasm, heterodimerized with antiapoptotic Bcl-2 family members.


Asunto(s)
Apoptosis/fisiología , Neutrófilos/fisiología , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/sangre , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas/sangre , Animales , Diferenciación Celular , División Celular , Línea Celular , Citoplasma/metabolismo , Humanos , Ratones , Ratones Noqueados , Mitocondrias/fisiología , Fosfatos/metabolismo , Fosforilación , Transporte de Proteínas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Proteína X Asociada a bcl-2
20.
Am J Respir Cell Mol Biol ; 26(4): 404-12, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11919076

RESUMEN

Prolonged eosinophil survival, i.e., reduced apoptosis, is implicated in the pathogenesis of chronic allergic inflammation. Here we demonstrate that interleukin (IL)-15, in the presence or absence of tumor necrosis factor (TNF)-alpha, reduces spontaneous apoptosis in freshly isolated human eosinophils. The prosurvival effect of IL-15 was abrogated by neutralizing antibody to granulocyte macrophage-colony stimulating factor (GM-CSF), although GM-CSF was not detected in conditioned media by ELISA. Additionally, the effect of IL-15 on spontaneous eosinophil apoptosis appeared to require nuclear factor-kappaB (NF-kappaB) activation based on evidence for NF-kappaB nuclear translocation and abrogation of the effect by the NF-kappaB inhibitor, Bay 11- 7082. Finally, the data demonstrate that IL-15 expression is higher in the submucosa of endobronchial tissues from subjects with moderate to severe asthma when compared with control subjects. Thus, our results suggest that IL-15, either alone or in combination with TNF-alpha, may perpetuate allergic inflammation by reduction of spontaneous eosinophil apoptosis through autocrine production of GM-CSF and NF-kappaB activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Eosinófilos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-15/metabolismo , FN-kappa B/metabolismo , Adolescente , Adulto , Apoptosis/fisiología , Asma/metabolismo , Asma/fisiopatología , Células Cultivadas , Eosinófilos/efectos de los fármacos , Eosinófilos/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de los fármacos , Humanos , Interleucina-15/farmacología , FN-kappa B/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA