Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Neurobiol Dis ; 159: 105492, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34478849

RESUMEN

Repulsive guidance molecule A (RGMa) is a potent inhibitor of axonal growth and a regulator of neuronal cell death. It is up-regulated following neuronal injury and accumulates in chronic neurodegenerative diseases. Neutralizing RGMa has the potential to promote neuroregeneration and neuroprotection. Previously we reported that a rat anti-N terminal RGMa (N-RGMa) antibody r5F9 and its humanized version h5F9 (ABT-207) promote neuroprotection and neuroregeneration in preclinical neurodegenerative disease models. However, due to its cross-reactivity to RGMc/hemojuvelin, ABT-207 causes iron accumulation in vivo, which could present a safety liability. Here we report the generation and characterization of a novel RGMa-selective anti-N-RGMa antibody elezanumab, which is currently under Phase 2 clinical evaluation in multiple disease indications. Elezanumab, a human monoclonal antibody generated by in vitro PROfusion mRNA display technology, competes with ABT-207 in binding to N-RGMa but lacks RGMc cross-reactivity with no impact on iron metabolism. It neutralizes repulsive activity of soluble RGMa in vitro and blocks membrane RGMa mediated BMP signaling. In the optic nerve crush and optic neuritis models, elezanumab promotes axonal regeneration and prevents retinal nerve fiber layer degeneration. In the spinal targeted experimental autoimmune encephalomyelitis (EAE) model, elezanumab promotes axonal regeneration and remyelination, decreases inflammatory lesion area and improves functional recovery. Finally, in the mouse cuprizone model, elezanumab reduces demyelination, which is consistent with its inhibitory effect on BMP signaling. Taken together, these preclinical data demonstrate that elezanumab has neuroregenerative and neuroprotective activities without impact on iron metabolism, thus providing a compelling rationale for its clinical development in neurodegenerative diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Proteínas Ligadas a GPI , Regeneración Nerviosa , Proteínas del Tejido Nervioso , Neuroprotección , Traumatismos del Nervio Óptico , Nervio Óptico , Neuritis Óptica , Recuperación de la Función , Retina , Animales , Ratones , Cuprizona/toxicidad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/fisiopatología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Inhibidores de la Monoaminooxidasa/toxicidad , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuroprotección/efectos de los fármacos , Nervio Óptico/efectos de los fármacos , Nervio Óptico/fisiología , Traumatismos del Nervio Óptico/fisiopatología , Neuritis Óptica/fisiopatología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Retina/efectos de los fármacos , Resonancia por Plasmón de Superficie
2.
Clin Transl Sci ; 17(1): e13706, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38266061

RESUMEN

Risankizumab is a high-affinity neutralizing anti-interleukin (IL)-23 monoclonal antibody marketed in over 40 countries across the globe to treat several inflammatory diseases, such as plaque psoriasis (PsO), psoriatic arthritis (PsA), and Crohn's disease (CD). This paper reviews the regulatory approval, mechanism of action, pharmacokinetics (PKs)/pharmacodynamics, immunogenicity, and clinical efficacy and safety data for risankizumab, focusing on the three main approved indications. Risankizumab binds to the p19 subunit of IL-23 and inhibits IL-23 from interacting with the IL-23 receptor and subsequent signaling. Biomarker data obtained following treatment with risankizumab in multiple indications provided supportive evidence for downstream blockade of IL-23 signaling associated with disease pathology. The PKs of risankizumab is linear and time-independent, consistent with typical IgG1 monoclonal antibodies, across all evaluated indications. Risankizumab exhibited positive exposure-response relationships for efficacy with no apparent exposure-dependent worsening in safety. Immunogenicity to risankizumab had no major clinical consequences for either efficacy or safety. Efficacy and safety of risankizumab have been established in PsO, PsA, and CD in the pivotal clinical trials where superior benefit/risk profiles were demonstrated compared to placebo and/or active comparators. Moreover, safety evaluations in open-label extension studies following long-term treatment with risankizumab showed stable and favorable safety profiles consistent with shorter-term studies. These data formed the foundation for risankizumab's marketing approvals to treat multiple inflammatory diseases across the globe.


Asunto(s)
Artritis Psoriásica , Enfermedad de Crohn , Humanos , Ciencia Traslacional Biomédica , Artritis Psoriásica/tratamiento farmacológico , Anticuerpos Monoclonales/efectos adversos , Interleucina-23
3.
JCI Insight ; 9(12)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38781017

RESUMEN

DC inhibitory receptor (DCIR) is a C-type lectin receptor selectively expressed on myeloid cells, including monocytes, macrophages, DCs, and neutrophils. Its role in immune regulation has been implicated in murine models and human genome-wide association studies, suggesting defective DCIR function associates with increased susceptibility to autoimmune diseases such as rheumatoid arthritis, lupus, and Sjögren's syndrome. However, little is known about the mechanisms underlying DCIR activation to dampen inflammation. Here, we developed anti-DCIR agonistic antibodies that promote phosphorylation on DCIR's immunoreceptor tyrosine-based inhibitory motifs and recruitment of SH2 containing protein tyrosine phosphatase-2 for reducing inflammation. We also explored the inflammation resolution by depleting DCIR+ cells with antibodies. Utilizing a human DCIR-knock-in mouse model, we validated the antiinflammatory properties of the agonistic anti-DCIR antibody in experimental peritonitis and colitis. These findings provide critical evidence for targeting DCIR to develop transformative therapies for inflammatory diseases.


Asunto(s)
Inflamación , Transducción de Señal , Animales , Ratones , Humanos , Transducción de Señal/inmunología , Inflamación/inmunología , Peritonitis/inmunología , Modelos Animales de Enfermedad , Colitis/inmunología , Fosforilación , Ratones Endogámicos C57BL
4.
Front Immunol ; 15: 1293883, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38455057

RESUMEN

Fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF) and systemic scleroderma (SSc), are commonly associated with high morbidity and mortality, thereby representing a significant unmet medical need. Interleukin 11 (IL11)-mediated cell activation has been identified as a central mechanism for promoting fibrosis downstream of TGFß. IL11 signaling has recently been reported to promote fibroblast-to-myofibroblast transition, thus leading to various pro-fibrotic phenotypic changes. We confirmed increased mRNA expression of IL11 and IL11Rα in fibrotic diseases by OMICs approaches and in situ hybridization. However, the vital role of IL11 as a driver for fibrosis was not recapitulated. While induction of IL11 secretion was observed downstream of TGFß signaling in human lung fibroblasts and epithelial cells, the cellular responses induced by IL11 was quantitatively and qualitatively inferior to that of TGFß at the transcriptional and translational levels. IL11 blocking antibodies inhibited IL11Rα-proximal STAT3 activation but failed to block TGFß-induced profibrotic signals. In summary, our results challenge the concept of IL11 blockade as a strategy for providing transformative treatment for fibrosis.


Asunto(s)
Interleucina-11 , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Fibrosis , Miofibroblastos/metabolismo
5.
Antib Ther ; 6(2): 137-146, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37342671

RESUMEN

The emergence of deep learning models such as AlphaFold2 has revolutionized the structure prediction of proteins. Nevertheless, much remains unexplored, especially on how we utilize structure models to predict biological properties. Herein, we present a method using features extracted from protein language models (PLMs) to predict the major histocompatibility complex class II (MHC-II) binding affinity of peptides. Specifically, we evaluated a novel transfer learning approach where the backbone of our model was interchanged with architectures designed for image classification tasks. Features extracted from several PLMs (ESM1b, ProtXLNet or ProtT5-XL-UniRef) were passed into image models (EfficientNet v2b0, EfficientNet v2m or ViT-16). The optimal pairing of the PLM and image classifier resulted in the final model TransMHCII, outperforming NetMHCIIpan 3.2 and NetMHCIIpan 4.0-BA on the receiver operating characteristic area under the curve, balanced accuracy and Jaccard scores. The architecture innovation may facilitate the development of other deep learning models for biological problems.

6.
MAbs ; 13(1): 1964420, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34460338

RESUMEN

Four antibodies that inhibit interleukin (IL)-23 are approved for the treatment of moderate-to-severe plaque psoriasis. Here, we present non-clinical data comparing ustekinumab, guselkumab, tildrakizumab and risankizumab with regard to thermostability, IL-23 binding affinity, inhibitory-binding mode, in vitro potency and in vivo efficacy. Risankizumab and guselkumab exhibited 5-fold higher affinity for IL-23 and showed more potent inhibition of IL-23 signaling than ustekinumab and tildrakizumab. Risankizumab and guselkumab completely blocked the binding of IL-23 to IL-23Rα as expected, whereas tildrakizumab did not. In vitro, risankizumab and guselkumab blocked the terminal differentiation of TH17 cells in a similar manner, while tildrakizumab had minimal impact on TH17 differentiation. In a human IL-23-induced ear-swelling mouse model, risankizumab and guselkumab were more effective than ustekinumab and tildrakizumab at reducing IL-17, IL-22, and keratinocyte gene expression. Our results indicate that the four clinically approved antibodies targeting IL-23 differ in affinity and binding epitope. These attributes contribute to differences in in vitro potency, receptor interaction inhibition mode and in vivo efficacy in preclinical studies as described in this report, and similarly may affect the clinical performance of these drugs.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Interleucina-23/antagonistas & inhibidores , Psoriasis/tratamiento farmacológico , Ustekinumab/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/metabolismo , Afinidad de Anticuerpos , Sitios de Unión de Anticuerpos , Células Cultivadas , Modelos Animales de Enfermedad , Estabilidad de Medicamentos , Epítopos , Femenino , Calor , Humanos , Interleucina-23/inmunología , Interleucina-23/metabolismo , Ratones Endogámicos C57BL , Desnaturalización Proteica , Estabilidad Proteica , Psoriasis/inmunología , Psoriasis/metabolismo , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Ustekinumab/inmunología , Ustekinumab/metabolismo
7.
Cell Rep ; 37(2): 109814, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34599871

RESUMEN

Control of the ongoing SARS-CoV-2 pandemic is endangered by the emergence of viral variants with increased transmission efficiency, resistance to marketed therapeutic antibodies, and reduced sensitivity to vaccine-induced immunity. Here, we screen B cells from COVID-19 donors and identify P5C3, a highly potent and broadly neutralizing monoclonal antibody with picomolar neutralizing activity against all SARS-CoV-2 variants of concern (VOCs) identified to date. Structural characterization of P5C3 Fab in complex with the spike demonstrates a neutralizing activity defined by a large buried surface area, highly overlapping with the receptor-binding domain (RBD) surface necessary for ACE2 interaction. We further demonstrate that P5C3 shows complete prophylactic protection in the SARS-CoV-2-infected hamster challenge model. These results indicate that P5C3 opens exciting perspectives either as a prophylactic agent in immunocompromised individuals with poor response to vaccination or as combination therapy in SARS-CoV-2-infected individuals.


Asunto(s)
Anticuerpos ampliamente neutralizantes/uso terapéutico , Tratamiento Farmacológico de COVID-19 , SARS-CoV-2/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Anticuerpos ampliamente neutralizantes/inmunología , COVID-19/inmunología , Línea Celular , Cricetinae , Modelos Animales de Enfermedad , Epítopos/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/metabolismo , Pruebas de Neutralización , Unión Proteica/inmunología , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Relación Estructura-Actividad , Vacunación
8.
N Engl J Med ; 351(20): 2069-79, 2004 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-15537905

RESUMEN

BACKGROUND: Crohn's disease is associated with excess cytokine activity mediated by type 1 helper T (Th1) cells. Interleukin-12 is a key cytokine that initiates Th1-mediated inflammatory responses. METHODS: This double-blind trial evaluated the safety and efficacy of a human monoclonal antibody against interleukin-12 (anti-interleukin-12) in 79 patients with active Crohn's disease. Patients were randomly assigned to receive seven weekly subcutaneous injections of 1 mg or 3 mg of anti-interleukin-12 per kilogram of body weight or placebo, with either a four-week interval between the first and second injection (Cohort 1) or no interruption between the two injections (Cohort 2). Safety was the primary end point, and the rates of clinical response (defined by a reduction in the score for the Crohn's Disease Activity Index [CDAI] of at least 100 points) and remission (defined by a CDAI score of 150 or less) were secondary end points. RESULTS: Seven weeks of uninterrupted treatment with 3 mg of anti-interleukin-12 per kilogram resulted in higher response rates than did placebo administration (75 percent vs. 25 percent, P=0.03). At 18 weeks of follow-up, the difference in response rates was no longer significant (69 percent vs. 25 percent, P=0.08). Differences in remission rates between the group given 3 mg of anti-interleukin-12 per kilogram and the placebo group in Cohort 2 were not significant at either the end of treatment or the end of follow-up (38 percent and 0 percent, respectively, at both times; P=0.07). There were no significant differences in response rates among the groups in Cohort 1. The rates of adverse events among patients receiving anti-interleukin-12 were similar to those among patients given placebo, except for a higher rate of local reactions at injection sites in the former group. Decreases in the secretion of interleukin-12, interferon-gamma, and tumor necrosis factor alpha by mononuclear cells of the colonic lamina propria accompanied clinical improvement in patients receiving anti-interleukin-12. CONCLUSIONS: Treatment with a monoclonal antibody against interleukin-12 may induce clinical responses and remissions in patients with active Crohn's disease. This treatment is associated with decreases in Th1-mediated inflammatory cytokines at the site of disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad de Crohn/tratamiento farmacológico , Interleucina-12/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Colon/inmunología , Enfermedad de Crohn/inmunología , Citocinas/metabolismo , Método Doble Ciego , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Inducción de Remisión
9.
Cell Rep ; 10(11): 1887-98, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25801027

RESUMEN

Repulsive guidance molecule A (RGMa) is a potent inhibitor of neuronal regeneration and a regulator of cell death, and it plays a role in multiple sclerosis (MS). In autopsy material from progressive MS patients, RGMa was found in active and chronic lesions, as well as in normal-appearing gray and white matter, and was expressed by cellular meningeal infiltrates. Levels of soluble RGMa in the cerebrospinal fluid were decreased in progressive MS patients successfully treated with intrathecal corticosteroid triamcinolone acetonide (TCA), showing functional improvements. In vitro, RGMa monoclonal antibodies (mAbs) reversed RGMa-mediated neurite outgrowth inhibition and chemorepulsion. In animal models of CNS damage and MS, RGMa antibody stimulated regeneration and remyelination of damaged nerve fibers, accelerated functional recovery, and protected the retinal nerve fiber layer as measured by clinically relevant optic coherence tomography. These data suggest that targeting RGMa is a promising strategy to improve functional recovery in MS patients.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Regeneración Nerviosa , Proteínas del Tejido Nervioso/metabolismo , Adulto , Anciano , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Femenino , Proteínas Ligadas a GPI , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Neuritas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/fisiología , Ratas , Ratas Wistar
10.
Immunol Lett ; 83(2): 77-83, 2002 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-12067755

RESUMEN

Antagonists of the B7 family of co-stimulatory molecules have the potential for altering immune responses therapeutically. To better define the requirements for such inhibitors, we have mapped the binding of an entire panel of blocking antibodies specific for human B7.1. By mutagenesis, each of the residues critical for blocking antibody binding appeared to fall entirely within the N-terminal V-set domain of B7.1. Thus, although antibody-antigen interacting surfaces can be quite large, these results indicate that a relatively small portion of the GFCC'C" face of this domain is crucial for further antagonist development.


Asunto(s)
Antígeno B7-1/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/genética , Anticuerpos Bloqueadores/inmunología , Antígeno B7-1/genética , Células COS , Mapeo Epitopo , Epítopos de Linfocito B , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
11.
Int Immunopharmacol ; 4(5): 693-708, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15120653

RESUMEN

Interleukin 22 (IL-22) is a cytokine induced during both innate and adaptive immune responses. It can effect an acute phase response, implicating a role for IL-22 in mechanisms of inflammation. IL-22 requires the presence of the IL-22 receptor (IL-22R) and IL-10 receptor 2 (IL-10R2) chains, two members of the class II cytokine receptor family (CRF2), to effect signal transduction within a cell. We studied the interaction between human IL-22 and the extracellular domains (ECD) of its receptor chains in an enzyme-linked immunoabsorbant assay (ELISA)-based format, using biotinylated IL-22 (bio-IL-22) and receptor-fusions containing the ECD of a receptor fused to the Fc of hIgG1 (IL-22R-Fc and IL-10R2-Fc). IL-22 has measurable affinity for IL-22R-Fc homodimer and undetectable affinity for IL-10R2. IL-22 has substantially greater affinity for IL-22R/IL-10R2-Fc heterodimers. Further analyses involving sequential additions of receptor homodimers and cytokine indicates that the IL-10R2(ECD) binds to a surface created by the interaction between IL-22 and the IL-22R(ECD), and thereby further stabilizes the association of IL-22 within this cytokine-receptor-Fc complex. Both a neutralizing rat monoclonal antibody, specific for human IL-22, and human IL-22BP-Fc, an Fc-fusion of the secreted IL-22 binding-protein and proposed natural antagonist for IL-22, bind to similar cytokine epitopes that may overlap the binding site for IL-22R(ECD). Another rat monoclonal antibody, specific for IL-22, binds to an epitope that may overlap a separate binding site for IL-10R2(ECD). We propose, based on this data, a temporal model for the development of a functional IL-22 cytokine-receptor complex.


Asunto(s)
Interleucinas/metabolismo , Receptores de Interleucina/metabolismo , Animales , Células CHO , Cricetinae , Dimerización , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Interleucinas/farmacología , Receptores de Interleucina/efectos de los fármacos , Receptores de Interleucina-10 , Factores de Tiempo , Interleucina-22
12.
Alzheimers Res Ther ; 6(3): 31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25024748

RESUMEN

INTRODUCTION: Immunotherapy targeting amyloid-ß peptide is under active clinical investigation for treatment of Alzheimer's disease (AD). Among the hypotheses being investigated for impact on clinical outcome are the preferred epitope or conformation of amyloid-ß to target for treatment, and the mechanism of action underlying immunotherapy. Bapineuzumab (humanized 3D6), a neo-epitope specific antibody recognizing amyloid-ß1-5 with strong preference for an exposed Asp residue at the N-terminus of the peptide, has undergone advanced clinical testing for treatment of AD. METHODS: To gain further insight into the epitope conformation, we interrogated structural details of amino-terminal epitopes in amyloid-ß using x-ray crystallography of 3D6Fab:amyloid-ß complexes. Humanization of 3D6 was carried out using standard procedures integrating recombinant methods, sequence informatics, and homology modeling predictions to identify important mouse framework residues for retention in the finished humanized product. RESULTS: Here we report the crystal structure of a recombinant Fab fragment of 3D6 in complex with amyloid-ß1-7 solved at 2.0 Å resolution. The N-terminus of amyloid-ß is bound to 3D6 as a 310 helix. The amino-terminal Asp residue is buried deepest in the antibody binding pocket, with the Cß atom of residue 6 visible at the entrance to the binding pocket near the surface of the antibody. We further evaluate homology model based predictions used to guide humanization of 3D6 to bapineuzumab, with actual structure of the Fab. The structure of the Fab:amyloid-ß complex validates design of the humanized antibody, and confirms the amyloid-ß epitope recognized by 3D6 as previously mapped by ELISA. CONCLUSIONS: The conformation of amyloid-ß antigen recognized by 3D6 is novel and distinct from other antibodies recognizing N-terminal epitopes. Our result provides the first report demonstrating structural conservation of antigen contact residues, and conformation of antigen recognized, between the parent murine antibody and its humanized version.

14.
Gastroenterology ; 122(1): 94-105, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11781285

RESUMEN

BACKGROUND & AIMS: Progression from the acute to chronic phase of inflammatory bowel disease cannot be easily evaluated in patients and has not been characterized in animal models. We report a longitudinal study investigating changes in the mucosal immune response in an experimental model of colitis. METHODS: Severity of colitis, body mass, stool consistency and blood content, serum amyloid A, and tissue histology were examined in interleukin (IL)-10-deficient mice over 35 weeks. The corresponding production of IL-12, IL-18, interferon gamma, tumor necrosis factor alpha, IL-4, and IL-13 by lamina propria mononuclear cells in the inflamed intestine was measured. Administration of neutralizing antibody to IL-12 at distinct times during disease progression permitted evaluation of its therapeutic potential. RESULTS: The clinical manifestations and intestinal inflammation delineated an early phase of colitis (10-24 weeks), characterized by a progressive increase in disease severity, followed by a late phase (>25 weeks), in which chronic inflammation persisted indefinitely. Lamina propria mononuclear cells from mice with early disease synthesized progressively greater quantities of IL-12 and interferon gamma, whereas production of both cytokines dramatically declined and returned to pre-disease levels in the late phase of colitis. Consistent with this pattern, neutralizing antibody to IL-12 reversed early, but not late, disease. In contrast, IL-4 and IL-13 production increased progressively from pre- to early to late disease. CONCLUSIONS: Colitis that develops in IL-10-deficient mice evolves into 2 distinct phases. IL-12 plays a pivotal role in early colitis, whereas its absence and the synthesis of IL-4 and IL-13 in late disease indicate that other immune mechanisms sustain chronic inflammation.


Asunto(s)
Colitis/inmunología , Interleucina-10/genética , Receptores de Interleucina/biosíntesis , Enfermedad Aguda , Animales , Anticuerpos/farmacología , Apolipoproteínas/metabolismo , Células Cultivadas , Enfermedad Crónica , Colitis/metabolismo , Diarrea/inmunología , Diarrea/metabolismo , Modelos Animales de Enfermedad , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-1/biosíntesis , Interleucina-13/biosíntesis , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-4/biosíntesis , Leucocitos/citología , Leucocitos/inmunología , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sangre Oculta , ARN Mensajero/análisis , Receptores de Interleucina/inmunología , Receptores de Interleucina-12 , Proteína Amiloide A Sérica/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
15.
J Immunol ; 170(2): 711-8, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12517932

RESUMEN

The program death 1 (PD-1) receptor and its ligands, PD-1 ligand (PD-L)1 and PD-L2, define a novel regulatory pathway with potential inhibitory effects on T, B, and monocyte responses. In the present study, we show that human CD4(+) T cells express PD-1, PD-L1, and PD-L2 upon activation, and Abs to the receptor can be agonists or antagonists of the pathway. Under optimal conditions of stimulation, ICOS but not CD28 costimulation can be prevented by PD-1 engagement. IL-2 levels induced by costimulation are critical in determining the outcome of the PD-1 engagement. Thus, low to marginal IL-2 levels produced upon ICOS costimulation account for the greater sensitivity of this pathway to PD-1-mediated inhibition. Interestingly, exogenous IL-2, IL-7, and IL-15 but not IL-4 and IL-21 can rescue PD-1 inhibition, suggesting that among these cytokines only those that activate STAT5 can rescue PD-1 inhibition. As STAT5 has been implicated in the maintenance of IL-2Ralpha expression, these results suggest that IL-7 and IL-15 restore proliferation under conditions of PD-1 engagement by enhancing high-affinity IL-2R expression and hence, IL-2 responsiveness.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/biosíntesis , Antígenos de Superficie/fisiología , Antígeno B7-1 , Antígenos CD28/biosíntesis , Citocinas/fisiología , Interleucina-15/biosíntesis , Interleucina-4/biosíntesis , Interleucina-7/biosíntesis , Interleucinas/biosíntesis , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Anticuerpos/fisiología , Antígenos CD , Antígenos de Diferenciación de Linfocitos T/fisiología , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis , Antígeno B7-H1 , Proteínas Sanguíneas/biosíntesis , Proteínas Sanguíneas/fisiología , División Celular/inmunología , Células Cultivadas , Regulación hacia Abajo/inmunología , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles , Péptidos y Proteínas de Señalización Intercelular , Subunidad gamma Común de Receptores de Interleucina , Interleucina-2/fisiología , Interleucina-7/fisiología , Glicoproteínas de Membrana , Péptidos/fisiología , Proteína 2 Ligando de Muerte Celular Programada 1 , Receptor de Muerte Celular Programada 1 , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-7/fisiología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Biochem Biophys Res Commun ; 300(4): 965-71, 2003 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-12559968

RESUMEN

A human therapeutic that specifically modulates skeletal muscle growth would potentially provide a benefit for a variety of conditions including sarcopenia, cachexia, and muscular dystrophy. Myostatin, a member of the TGF-beta family of growth factors, is a known negative regulator of muscle mass, as mice lacking the myostatin gene have increased muscle mass. Thus, an inhibitor of myostatin may be useful therapeutically as an anabolic agent for muscle. However, since myostatin is expressed in both developing and adult muscles, it is not clear whether it regulates muscle mass during development or in adults. In order to test the hypothesis that myostatin regulates muscle mass in adults, we generated an inhibitory antibody to myostatin and administered it to adult mice. Here we show that mice treated pharmacologically with an antibody to myostatin have increased skeletal muscle mass and increased grip strength. These data show for the first time that myostatin acts postnatally as a negative regulator of skeletal muscle growth and suggest that myostatin inhibitors could provide a therapeutic benefit in diseases for which muscle mass is limiting.


Asunto(s)
Músculo Esquelético/crecimiento & desarrollo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Peso Corporal , Células CHO , Cricetinae , Medios de Cultivo Condicionados , Femenino , Fuerza de la Mano , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/anatomía & histología , Músculo Esquelético/fisiología , Miostatina , Unión Proteica , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA