Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Biol Chem ; 295(16): 5404-5418, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32075906

RESUMEN

Myostatin (or growth/differentiation factor 8 (GDF8)) is a member of the transforming growth factor ß superfamily of growth factors and negatively regulates skeletal muscle growth. Its dysregulation is implicated in muscle wasting diseases. SRK-015 is a clinical-stage mAb that prevents extracellular proteolytic activation of pro- and latent myostatin. Here we used integrated structural and biochemical approaches to elucidate the molecular mechanism of antibody-mediated neutralization of pro-myostatin activation. The crystal structure of pro-myostatin in complex with 29H4-16 Fab, a high-affinity variant of SRK-015, at 2.79 Å resolution revealed that the antibody binds to a conformational epitope in the arm region of the prodomain distant from the proteolytic cleavage sites. This epitope is highly sequence-divergent, having only limited similarity to other closely related members of the transforming growth factor ß superfamily. Hydrogen/deuterium exchange MS experiments indicated that antibody binding induces conformational changes in pro- and latent myostatin that span the arm region, the loops contiguous to the protease cleavage sites, and the latency-associated structural elements. Moreover, negative-stain EM with full-length antibodies disclosed a stable, ring-like antigen-antibody structure in which the two Fab arms of a single antibody occupy the two arm regions of the prodomain in the pro- and latent myostatin homodimers, suggesting a 1:1 (antibody:myostatin homodimer) binding stoichiometry. These results suggest that SRK-015 binding stabilizes the latent conformation and limits the accessibility of protease cleavage sites within the prodomain. These findings shed light on approaches that specifically block the extracellular activation of growth factors by targeting their precursor forms.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Simulación del Acoplamiento Molecular , Miostatina/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Sitios de Unión , Humanos , Miostatina/antagonistas & inhibidores , Miostatina/inmunología , Unión Proteica , Estabilidad Proteica
2.
J Autoimmun ; 124: 102726, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34555678

RESUMEN

The ability of regulatory T (Treg) cells to migrate into inflammatory sites is reduced in autoimmune diseases, including rheumatoid arthritis (RA). The reasons for impaired Treg cell migration remain largely unknown. We performed multiplex human kinase activity arrays to explore possible differences in the post-translational phosphorylation status of kinase related proteins that could account for altered Treg cell migration in RA. Results were verified by migration assays and Western blot analysis of CD4+ T cells from RA patients and from mice with collagen type II induced arthritis. Kinome profiling of CD4+ T cells from RA patients revealed significantly altered post-translational phosphorylation of kinase related proteins, including G-protein-signaling modulator 2 (GPSM2), protein tyrosine kinase 6 (PTK6) and vitronectin precursor (VTNC). These proteins have not been associated with RA until now. We found that GPSM2 expression is reduced in CD4+ T cells from RA patients and is significantly downregulated in experimental autoimmune arthritis following immunization of mice with collagen type II. Interestingly, GPSM2 acts as a promoter of Treg cell migration in healthy individuals. Treatment of RA patients with interleukin-6 receptor (IL-6R) blocking antibodies restores GPSM2 expression, thereby improving Treg cell migration. Our study highlights the potential of multiplex kinase activity arrays as a tool for the identification of RA-related proteins which could serve as targets for novel treatments.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Bloqueadores/metabolismo , Movimiento Celular , Células Cultivadas , Colágeno Tipo II/inmunología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos DBA , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores de Interleucina-6/inmunología
3.
mBio ; : e0250623, 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37937842

RESUMEN

Lipid nanoparticle (LNP)-encapsulated mRNAs have emerged as effective vaccination tools to stimulate immunity. The most common application of this technology is to deliver mRNAs that encode antigenic proteins to dendritic cells (DCs), which then stimulate antigen-specific lymphocyte responses. It is unclear whether other immunostimulatory DC activities necessary for vaccine efficacy, beyond antigen presentation, can be induced via mRNA-encoded proteins. Herein, we report an mRNA encoding a self-DNA reactive variant of the enzyme cyclic GMP-AMP synthase (cGAS), known as cGAS∆N. cGAS∆N produces the cyclic dinucleotide cGAMP upon binding intra-mitochondrial DNA. cGAMP binds the protein STING, which activates innate immune responses that stimulate T cells. We found that when delivered to DCs via LNPs, mRNA-encoded cGAS∆N induced the upregulation of chemokine receptors, T cell costimulatory molecules, major histocompatibility complex proteins, pro-inflammatory cytokines and type I interferons from murine and human DCs. These activities exceeded the immunostimulatory activities of mRNA-encoded antigens delivered via LNPs. Co-immunization of mice with antigen-LNPs and cGAS∆N-LNPs led to the robust production of antigen-specific IFNγ-producing T cells. These T cell responses were durable and circulated through the lymphatics, blood, and lungs. Immunizations with antigen-LNPs alone, akin to what are used in the clinic, stimulated weak and transient T cell responses. Antibody responses to antigen-LNPs were biased towards type I isotypes when co-injected with cGAS∆N-LNPs, as compared to immunizations with antigen-LNPs alone. These findings establish the enzyme cGAS∆N as a catalytic adjuvant, which may prove useful in enhancing the immunogenicity of nucleic acid-based vaccines. IMPORTANCE Nucleic acid-based vaccines hold promise in preventing infections and treating cancer. The most common use of this technology is to encode antigenic proteins on mRNAs that are delivered to cells via lipid nanoparticle (LNP) formulations. In this study, we discovered that immunostimulatory proteins can also be encoded on mRNAs in LNPs. We found that an active mutant of the enzyme cGAS, referred to as cGAS∆N, acts as a catalytic adjuvant in LNP-encapsulated mRNA vaccines. The delivery of cGAS∆N mRNA via LNPs in combination with antigen mRNA-LNPs led to durable antigen-specific IFNγ-producing T cells that exceeded the efficiency of antigen-LNPs similar to those currently used in the clinic. This strategy did not compromise B cell responses; rather it induced Th1-biased antibody isotypes. This work unveils new vaccine design strategies using mRNA-encoded catalytic adjuvants that could be ideal for generating CD8+ T cell and B cell responses for immunotherapies.

4.
Physiol Rep ; 11(19): e15829, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37771106

RESUMEN

The JAK/STAT pathway plays a crucial role in the pathogenesis of rheumatoid arthritis (RA) and JAK inhibitors have emerged as a new group of effective drugs for RA treatment. Recently, high STAT3 levels have been associated with the upregulation of the scaffold protein NEDD9, which is a regulator of T-cell trafficking and promotes collagen-induced arthritis (CIA). In this study, we aimed to reveal how treatment with JAK inhibitors affects NEDD9 in CD4+ T cells from RA patients. We analyzed NEDD9 expression in CD4+ T cells from 50 patients treated with either baricitinib, tofacitinib, or upadacitinib and performed cell migration assays to assess the potential influence of JAK inhibitor treatment on CD4+ T-cell migration. We observed that treatment with baricitinib and upadacitinib is associated with reduced NEDD9 expression in CD4+ T cells. In contrast, NEDD9 levels were not altered during treatment with tofacitinib. Moreover, treatment with baricitinib was associated with a significantly reduced migratory capacity of effector CD4+ T cells but not with impaired migration of Treg cells. This study reveals previously unknown associations between JAK inhibitor treatment and NEDD9 expression and indicates that JAK inhibitors could reduce effector T-cell migration.


Asunto(s)
Artritis Reumatoide , Inhibidores de las Cinasas Janus , Humanos , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus , Linfocitos T CD4-Positivos/patología , Transducción de Señal , Factores de Transcripción STAT , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Proteínas Adaptadoras Transductoras de Señales
5.
Leukemia ; 36(7): 1794-1805, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35523865

RESUMEN

The scaffold protein NEDD9 is frequently upregulated and hyperphosphorylated in cancers, and is associated with poor clinical outcome. NEDD9 promotes B-cell adhesion, migration and chemotaxis, pivotal processes for malignant development. We show that global or B-cell-specific deletion of Nedd9 in chronic lymphocytic leukemia (CLL) mouse models delayed CLL development, markedly reduced disease burden and resulted in significant survival benefit. NEDD9 was required for efficient CLL cell homing, chemotaxis, migration and adhesion. In CLL patients, peripheral NEDD9 expression was associated with adhesion and migration signatures as well as leukocyte count. Additionally, CLL lymph nodes frequently expressed high NEDD9 levels, with a subset of patients showing NEDD9 expression enriched in the CLL proliferation centers. Blocking activity of prominent NEDD9 effectors, including AURKA and HDAC6, effectively reduced CLL cell migration and chemotaxis. Collectively, our study provides evidence for a functional role of NEDD9 in CLL pathogenesis that involves intrinsic defects in adhesion, migration and homing.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Leucemia Linfocítica Crónica de Células B , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Aurora Quinasa A , Movimiento Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Leucemia Linfocítica Crónica de Células B/patología , Ratones
6.
Sci Transl Med ; 12(536)2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32213632

RESUMEN

Despite breakthroughs achieved with cancer checkpoint blockade therapy (CBT), many patients do not respond to anti-programmed cell death-1 (PD-1) due to primary or acquired resistance. Human tumor profiling and preclinical studies in tumor models have recently uncovered transforming growth factor-ß (TGFß) signaling activity as a potential point of intervention to overcome primary resistance to CBT. However, the development of therapies targeting TGFß signaling has been hindered by dose-limiting cardiotoxicities, possibly due to nonselective inhibition of multiple TGFß isoforms. Analysis of mRNA expression data from The Cancer Genome Atlas revealed that TGFΒ1 is the most prevalent TGFß isoform expressed in many types of human tumors, suggesting that TGFß1 may be a key contributor to primary CBT resistance. To test whether selective TGFß1 inhibition is sufficient to overcome CBT resistance, we generated a high-affinity, fully human antibody, SRK-181, that selectively binds to latent TGFß1 and inhibits its activation. Coadministration of SRK-181-mIgG1 and an anti-PD-1 antibody in mice harboring syngeneic tumors refractory to anti-PD-1 treatment induced profound antitumor responses and survival benefit. Specific targeting of TGFß1 was also effective in tumors expressing more than one TGFß isoform. Combined SRK-181-mIgG1 and anti-PD-1 treatment resulted in increased intratumoral CD8+ T cells and decreased immunosuppressive myeloid cells. No cardiac valvulopathy was observed in a 4-week rat toxicology study with SRK-181, suggesting that selectively blocking TGFß1 activation may avoid dose-limiting toxicities previously observed with pan-TGFß inhibitors. These results establish a rationale for exploring selective TGFß1 inhibition to overcome primary resistance to CBT.


Asunto(s)
Neoplasias , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Linfocitos T CD8-positivos , Cardiotoxicidad , Línea Celular Tumoral , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Ratas , Transducción de Señal
7.
Sci Rep ; 6: 24650, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27090146

RESUMEN

Next generation sequencing is a transformative technology for discovering and diagnosing genetic disorders. However, high-throughput sequencing remains error-prone, necessitating variant confirmation in order to meet the exacting demands of clinical diagnostic sequencing. To address this, we devised an orthogonal, dual platform approach employing complementary target capture and sequencing chemistries to improve speed and accuracy of variant calls at a genomic scale. We combined DNA selection by bait-based hybridization followed by Illumina NextSeq reversible terminator sequencing with DNA selection by amplification followed by Ion Proton semiconductor sequencing. This approach yields genomic scale orthogonal confirmation of ~95% of exome variants. Overall variant sensitivity improves as each method covers thousands of coding exons missed by the other. We conclude that orthogonal NGS offers improvements in variant calling sensitivity when two platforms are used, better specificity for variants identified on both platforms, and greatly reduces the time and expense of Sanger follow-up, thus enabling physicians to act on genomic results more quickly.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Técnicas de Diagnóstico Molecular/métodos , Análisis de Secuencia de ADN/métodos , Exoma , Humanos , Reacción en Cadena de la Polimerasa/métodos , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA