Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Annu Rev Immunol ; 33: 139-67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25493332

RESUMEN

Cytokines exert a vast array of immunoregulatory actions critical to human biology and disease. However, the desired immunotherapeutic effects of native cytokines are often mitigated by toxicity or lack of efficacy, either of which results from cytokine receptor pleiotropy and/or undesired activation of off-target cells. As our understanding of the structural principles of cytokine-receptor interactions has advanced, mechanism-based manipulation of cytokine signaling through protein engineering has become an increasingly feasible and powerful approach. Modified cytokines, both agonists and antagonists, have been engineered with narrowed target cell specificities, and they have also yielded important mechanistic insights into cytokine biology and signaling. Here we review the theory and practice of cytokine engineering and rationalize the mechanisms of several engineered cytokines in the context of structure. We discuss specific examples of how structure-based cytokine engineering has opened new opportunities for cytokines as drugs, with a focus on the immunotherapeutic cytokines interferon, interleukin-2, and interleukin-4.


Asunto(s)
Citocinas/genética , Citocinas/metabolismo , Ingeniería Genética , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Animales , Citocinas/química , Espacio Extracelular/metabolismo , Humanos , Espacio Intracelular/metabolismo , Unión Proteica , Transporte de Proteínas , Receptores de Citocinas/química , Transducción de Señal
2.
Cell ; 168(6): 1041-1052.e18, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283060

RESUMEN

Most secreted growth factors and cytokines are functionally pleiotropic because their receptors are expressed on diverse cell types. While important for normal mammalian physiology, pleiotropy limits the efficacy of cytokines and growth factors as therapeutics. Stem cell factor (SCF) is a growth factor that acts through the c-Kit receptor tyrosine kinase to elicit hematopoietic progenitor expansion but can be toxic when administered in vivo because it concurrently activates mast cells. We engineered a mechanism-based SCF partial agonist that impaired c-Kit dimerization, truncating downstream signaling amplitude. This SCF variant elicited biased activation of hematopoietic progenitors over mast cells in vitro and in vivo. Mouse models of SCF-mediated anaphylaxis, radioprotection, and hematopoietic expansion revealed that this SCF partial agonist retained therapeutic efficacy while exhibiting virtually no anaphylactic off-target effects. The approach of biasing cell activation by tuning signaling thresholds and outputs has applications to many dimeric receptor-ligand systems.


Asunto(s)
Anafilaxia/metabolismo , Células Madre Hematopoyéticas/inmunología , Mastocitos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo , Anafilaxia/inmunología , Animales , Dimerización , Humanos , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas c-kit/agonistas , Proteínas Proto-Oncogénicas c-kit/química , Factor de Células Madre/química , Factor de Células Madre/genética
3.
Cell ; 168(6): 1053-1064.e15, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283061

RESUMEN

Cytokines are classically thought to stimulate downstream signaling pathways through monotonic activation of receptors. We describe a severe anemia resulting from a homozygous mutation (R150Q) in the cytokine erythropoietin (EPO). Surprisingly, the EPO R150Q mutant shows only a mild reduction in affinity for its receptor but has altered binding kinetics. The EPO mutant is less effective at stimulating erythroid cell proliferation and differentiation, even at maximally potent concentrations. While the EPO mutant can stimulate effectors such as STAT5 to a similar extent as the wild-type ligand, there is reduced JAK2-mediated phosphorylation of select downstream targets. This impairment in downstream signaling mechanistically arises from altered receptor dimerization dynamics due to extracellular binding changes. These results demonstrate how variation in a single cytokine can lead to biased downstream signaling and can thereby cause human disease. Moreover, we have defined a distinct treatable form of anemia through mutation identification and functional studies.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/patología , Eritropoyetina/genética , Mutación Missense , Transducción de Señal , Anemia de Diamond-Blackfan/terapia , Niño , Consanguinidad , Activación Enzimática , Eritropoyesis , Eritropoyetina/química , Femenino , Humanos , Janus Quinasa 2/metabolismo , Cinética , Masculino , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo
4.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25728669

RESUMEN

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Asunto(s)
Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Línea Celular , Cristalografía por Rayos X , Dimerización , Eritropoyetina/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Mutación Puntual , Ingeniería de Proteínas , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/antagonistas & inhibidores , Alineación de Secuencia
5.
Immunity ; 46(3): 379-392, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28329704

RESUMEN

Type III interferons (IFN-λs) signal through a heterodimeric receptor complex composed of the IFN-λR1 subunit, specific for IFN-λs, and interleukin-10Rß (IL-10Rß), which is shared by multiple cytokines in the IL-10 superfamily. Low affinity of IL-10Rß for cytokines has impeded efforts aimed at crystallizing cytokine-receptor complexes. We used yeast surface display to engineer a higher-affinity IFN-λ variant, H11, which enabled crystallization of the ternary complex. The structure revealed that IL-10Rß uses a network of tyrosine residues as hydrophobic anchor points to engage IL-10 family cytokines that present complementary hydrophobic binding patches, explaining its role as both a cross-reactive but cytokine-specific receptor. H11 elicited increased anti-proliferative and antiviral activities in vitro and in vivo. In contrast, engineered higher-affinity type I IFNs did not increase antiviral potency over wild-type type I IFNs. Our findings provide insight into cytokine recognition by the IL-10R family and highlight the plasticity of type III interferon signaling and its therapeutic potential.


Asunto(s)
Interferones/inmunología , Receptores de Interferón/inmunología , Receptores de Interleucina-10/inmunología , Animales , Línea Celular , Cristalografía por Rayos X , Citometría de Flujo , Humanos , Ratones , Reacción en Cadena de la Polimerasa , Resonancia por Plasmón de Superficie
6.
Cell ; 146(4): 621-32, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21854986

RESUMEN

Type I Interferons (IFNs) are important cytokines for innate immunity against viruses and cancer. Sixteen human type I IFN variants signal through the same cell-surface receptors, IFNAR1 and IFNAR2, yet they can evoke markedly different physiological effects. The crystal structures of two human type I IFN ternary signaling complexes containing IFNα2 and IFNω reveal recognition modes and heterotrimeric architectures that are unique among the cytokine receptor superfamily but conserved between different type I IFNs. Receptor-ligand cross-reactivity is enabled by conserved receptor-ligand "anchor points" interspersed among ligand-specific interactions that "tune" the relative IFN-binding affinities, in an apparent extracellular "ligand proofreading" mechanism that modulates biological activity. Functional differences between IFNs are linked to their respective receptor recognition chemistries, in concert with a ligand-induced conformational change in IFNAR1, that collectively control signal initiation and complex stability, ultimately regulating differential STAT phosphorylation profiles, receptor internalization rates, and downstream gene expression patterns.


Asunto(s)
Interferón Tipo I/química , Interferón-alfa/química , Receptores de Interferón/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Interferón Tipo I/metabolismo , Interferón-alfa/metabolismo , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Alineación de Secuencia
7.
Proc Natl Acad Sci U S A ; 119(33): e2112006119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35939683

RESUMEN

IL13Rα2 is an attractive target due to its overexpression in a variety of cancers and rare expression in healthy tissue, motivating expansion of interleukin 13 (IL13)-based chimeric antigen receptor (CAR) T cell therapy from glioblastoma into systemic malignancies. IL13Rα1, the other binding partner of IL13, is ubiquitously expressed in healthy tissue, raising concerns about the therapeutic window of systemic administration. IL13 mutants with diminished binding affinity to IL13Rα1 were previously generated by structure-guided protein engineering. In this study, two such variants, termed C4 and D7, are characterized for their ability to mediate IL13Rα2-specific response as binding domains for CAR T cells. Despite IL13Rα1 and IL13Rα2 sharing similar binding interfaces on IL13, mutations to IL13 that decrease binding affinity for IL13Rα1 did not drastically change binding affinity for IL13Rα2. Micromolar affinity to IL13Rα1 was sufficient to pacify IL13-mutein CAR T cells in the presence of IL13Rα1-overexpressing cells in vitro. Interestingly, effector activity of D7 CAR T cells, but not C4 CAR T cells, was demonstrated when cocultured with IL13Rα1/IL4Rα-coexpressing cancer cells. While low-affinity interactions with IL13Rα1 did not result in observable toxicities in mice, in vivo biodistribution studies demonstrated that C4 and D7 CAR T cells were better able to traffic away from IL13Rα1+ lung tissue than were wild-type (WT) CAR T cells. These results demonstrate the utility of structure-guided engineering of ligand-based binding domains with appropriate selectivity while validating IL13-mutein CARs with improved selectivity for application to systemic IL13Rα2-expressing malignancies.


Asunto(s)
Inmunoterapia Adoptiva , Subunidad alfa2 del Receptor de Interleucina-13 , Interleucina-13 , Neoplasias , Animales , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-13/genética , Interleucina-13/farmacocinética , Interleucina-13/uso terapéutico , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Ratones , Neoplasias/terapia , Ingeniería de Proteínas , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Nat Immunol ; 13(12): 1187-95, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23104097

RESUMEN

Interleukin 15 (IL-15) and IL-2 have distinct immunological functions even though both signal through the receptor subunit IL-2Rß and the common γ-chain (γ(c)). Here we found that in the structure of the IL-15-IL-15Rα-IL-2Rß-γ(c) quaternary complex, IL-15 binds to IL-2Rß and γ(c) in a heterodimer nearly indistinguishable from that of the IL-2-IL-2Rα-IL-2Rß-γ(c) complex, despite their different receptor-binding chemistries. IL-15Rα substantially increased the affinity of IL-15 for IL-2Rß, and this allostery was required for IL-15 trans signaling. Consistent with their identical IL-2Rß-γ(c) dimer geometries, IL-2 and IL-15 showed similar signaling properties in lymphocytes, with any differences resulting from disparate receptor affinities. Thus, IL-15 and IL-2 induced similar signals, and the cytokine specificity of IL-2Rα versus IL-15Rα determined cellular responsiveness. Our results provide new insights for the development of specific immunotherapeutics based on IL-15 or IL-2.


Asunto(s)
Interleucina-15/inmunología , Interleucina-2/inmunología , Animales , Sitios de Unión , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Interleucina-15/química , Interleucina-15/metabolismo , Interleucina-2/química , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad beta del Receptor de Interleucina-2/metabolismo , Ligandos , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transducción de Señal
9.
EMBO Rep ; 23(10): e55450, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35920255

RESUMEN

Interleukin 27 (IL-27) is a heterodimeric cytokine that elicits potent immunosuppressive responses. Comprised of EBI3 and p28 subunits, IL-27 binds GP130 and IL-27Rα receptor chains to activate the JAK/STAT signaling cascade. However, how these receptors recognize IL-27 and form a complex capable of phosphorylating JAK proteins remains unclear. Here, we used cryo electron microscopy (cryoEM) and AlphaFold modeling to solve the structure of the IL-27 receptor recognition complex. Our data show how IL-27 serves as a bridge connecting IL-27Rα (domains 1-2) with GP130 (domains 1-3) to initiate signaling. While both receptors contact the p28 component of the heterodimeric cytokine, EBI3 stabilizes the complex by binding a positively charged surface of IL-27Rα and Domain 1 of GP130. We find that assembly of the IL-27 receptor recognition complex is distinct from both IL-12 and IL-6 cytokine families and provides a mechanistic blueprint for tuning IL-27 pleiotropic actions.


Asunto(s)
Receptor gp130 de Citocinas , Interleucina-27 , Receptores de Interleucina , Receptor gp130 de Citocinas/química , Humanos , Interleucina-12 , Interleucina-27/química , Interleucina-6 , Interleucinas , Receptores de Interleucina/química
10.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33384332

RESUMEN

Thrombopoietin (TPO) and the TPO-receptor (TPO-R, or c-MPL) are essential for hematopoietic stem cell (HSC) maintenance and megakaryocyte differentiation. Agents that can modulate TPO-R signaling are highly desirable for both basic research and clinical utility. We developed a series of surrogate protein ligands for TPO-R, in the form of diabodies (DBs), that homodimerize TPO-R on the cell surface in geometries that are dictated by the DB receptor binding epitope, in effect "tuning" downstream signaling responses. These surrogate ligands exhibit diverse pharmacological properties, inducing graded signaling outputs, from full to partial TPO agonism, thus decoupling the dual functions of TPO/TPO-R. Using single-cell RNA sequencing and HSC self-renewal assays we find that partial agonistic diabodies preserved the stem-like properties of cultured HSCs, but also blocked oncogenic colony formation in essential thrombocythemia (ET) through inverse agonism. Our data suggest that dampening downstream TPO signaling is a powerful approach not only for HSC preservation in culture, but also for inhibiting oncogenic signaling through the TPO-R.


Asunto(s)
Receptores de Trombopoyetina/metabolismo , Trombopoyetina/metabolismo , Diferenciación Celular/fisiología , Membrana Celular/metabolismo , Epítopos/inmunología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ligandos , Megacariocitos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Trombopoyetina/inmunología , Receptores de Trombopoyetina/fisiología , Transducción de Señal/fisiología , Trombocitemia Esencial/metabolismo , Trombopoyetina/fisiología
11.
PLoS Biol ; 17(11): e3000540, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31770366

RESUMEN

Interleukin-22 (IL-22) is a critical immune defence cytokine that maintains intestinal homeostasis and promotes wound healing and tissue regeneration, which can support the growth of colorectal tumours. Mutations in the adenomatous polyposis coli gene (Apc) are a major driver of familial colorectal cancers (CRCs). How IL-22 contributes to APC-mediated tumorigenesis is poorly understood. To investigate IL-22 signalling in wild-type (WT) and APC-mutant cells, we performed RNA sequencing (RNAseq) of IL-22-treated murine small intestinal epithelial organoids. In WT epithelia, antimicrobial defence and cellular stress response pathways were most strongly induced by IL-22. Surprisingly, although IL-22 activates signal transducer and activator of transcription 3 (STAT3) in APC-mutant cells, STAT3 target genes were not induced. Our analyses revealed that ApcMin/Min cells are resistant to IL-22 due to reduced expression of the IL-22 receptor, and increased expression of inhibitors of STAT3, particularly histone deacetylases (HDACs). We further show that IL-22 increases DNA damage and genomic instability, which can accelerate cellular transition from heterozygosity (ApcMin/+) to homozygosity (ApcMin/Min) to drive tumour formation. Our data reveal an unexpected role for IL-22 in promoting early tumorigenesis while excluding a function for IL-22 in transformed epithelial cells.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Células Epiteliales/metabolismo , Interleucinas/metabolismo , Poliposis Adenomatosa del Colon/genética , Animales , Carcinogénesis/genética , Neoplasias Colorrectales/metabolismo , Citocinas/metabolismo , Femenino , Interleucinas/genética , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Intestinos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo , Análisis de Secuencia de ARN/métodos , Transducción de Señal , Interleucina-22
12.
J Biol Chem ; 294(38): 13876-13886, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31387945

RESUMEN

Ligand-induced dimerization is the predominant mechanism through which secreted proteins activate cell surface receptors to transmit essential biological signals. Cytokines are a large class of soluble proteins that dimerize transmembrane receptors into precise signaling topologies, but there is a need for alternative, engineerable ligand scaffolds that specifically recognize and stabilize these protein interactions. Recombinant antibodies can potentially serve as robust and versatile platforms for cytokine complex stabilization, and their specificity allows for tunable modulation of dimerization equilibrium. Here, we devised an evolutionary strategy to isolate monovalent antibody fragments that bridge together two different receptor subunits in a cytokine-receptor complex, precisely as the receptors are disposed in their natural signaling orientations. To do this, we screened a naive antibody library against a stabilized ligand-receptor ternary complex that acted as a "molecular cast" of the natural receptor dimer conformation. Our selections elicited "stapler" single-chain variable fragments (scFvs) of antibodies that specifically engage the interleukin-4 receptor heterodimer. The 3.1 Å resolution crystal structure of one such stapler revealed that, as intended, this scFv recognizes a composite epitope between the two receptors as they are positioned in the complex. Extending our approach, we evolved a stapler scFv that specifically binds to and stabilizes the interface between the interleukin-2 cytokine and one of its receptor subunits, leading to a 15-fold enhancement in interaction affinity. This demonstration that scFvs can be selected to recognize epitopes that span protein interfaces presents new opportunities to engineer structurally defined antibodies for a broad range of research and therapeutic applications.


Asunto(s)
Epítopos/química , Ingeniería de Proteínas/métodos , Anticuerpos de Cadena Única/genética , Secuencia de Aminoácidos/genética , Dimerización , Mapeo Epitopo/métodos , Humanos , Ligandos , Conformación Molecular , Biblioteca de Péptidos , Unión Proteica/genética , Multimerización de Proteína/genética
13.
Nature ; 484(7395): 529-33, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446627

RESUMEN

The immunostimulatory cytokine interleukin-2 (IL-2) is a growth factor for a wide range of leukocytes, including T cells and natural killer (NK) cells. Considerable effort has been invested in using IL-2 as a therapeutic agent for a variety of immune disorders ranging from AIDS to cancer. However, adverse effects have limited its use in the clinic. On activated T cells, IL-2 signals through a quaternary 'high affinity' receptor complex consisting of IL-2, IL-2Rα (termed CD25), IL-2Rß and IL-2Rγ. Naive T cells express only a low density of IL-2Rß and IL-2Rγ, and are therefore relatively insensitive to IL-2, but acquire sensitivity after CD25 expression, which captures the cytokine and presents it to IL-2Rß and IL-2Rγ. Here, using in vitro evolution, we eliminated the functional requirement of IL-2 for CD25 expression by engineering an IL-2 'superkine' (also called super-2) with increased binding affinity for IL-2Rß. Crystal structures of the IL-2 superkine in free and receptor-bound forms showed that the evolved mutations are principally in the core of the cytokine, and molecular dynamics simulations indicated that the evolved mutations stabilized IL-2, reducing the flexibility of a helix in the IL-2Rß binding site, into an optimized receptor-binding conformation resembling that when bound to CD25. The evolved mutations in the IL-2 superkine recapitulated the functional role of CD25 by eliciting potent phosphorylation of STAT5 and vigorous proliferation of T cells irrespective of CD25 expression. Compared to IL-2, the IL-2 superkine induced superior expansion of cytotoxic T cells, leading to improved antitumour responses in vivo, and elicited proportionally less expansion of T regulatory cells and reduced pulmonary oedema. Collectively, we show that in vitro evolution has mimicked the functional role of CD25 in enhancing IL-2 potency and regulating target cell specificity, which has implications for immunotherapy.


Asunto(s)
Evolución Molecular Dirigida , Interleucina-2/química , Interleucina-2/inmunología , Proteínas Mutantes/química , Proteínas Mutantes/inmunología , Ingeniería de Proteínas , Animales , Sitios de Unión , Línea Celular , Proliferación Celular , Cristalografía por Rayos X , Humanos , Inmunoterapia , Interleucina-2/genética , Interleucina-2/farmacología , Subunidad alfa del Receptor de Interleucina-2/química , Subunidad alfa del Receptor de Interleucina-2/deficiencia , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad beta del Receptor de Interleucina-2/química , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Simulación de Dinámica Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/farmacología , Mutación , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Fosforilación , Conformación Proteica , Factor de Transcripción STAT5/metabolismo , Resonancia por Plasmón de Superficie , Linfocitos T/citología , Linfocitos T/inmunología
14.
Nat Chem Biol ; 8(12): 990-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23103943

RESUMEN

Cytokines dimerize their receptors, with the binding of the 'second chain' triggering signaling. In the interleukin (IL)-4 and IL-13 system, different cell types express varying numbers of alternative second receptor chains (γc or IL-13Rα1), forming functionally distinct type I or type II complexes. We manipulated the affinity and specificity of second chain recruitment by human IL-4. A type I receptor-selective IL-4 'superkine' with 3,700-fold higher affinity for γc was three- to ten-fold more potent than wild-type IL-4. Conversely, a variant with high affinity for IL-13Rα1 more potently activated cells expressing the type II receptor and induced differentiation of dendritic cells from monocytes, implicating the type II receptor in this process. Superkines showed signaling advantages on cells with lower second chain numbers. Comparative transcriptional analysis reveals that the superkines induce largely redundant gene expression profiles. Variable second chain numbers can be exploited to redirect cytokines toward distinct cell subsets and elicit new actions, potentially improving the selectivity of cytokine therapy.


Asunto(s)
Citocinas/fisiología , Interleucina-4/análogos & derivados , Interleucina-4/farmacología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Dendríticas/efectos de los fármacos , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Factores Inmunológicos/farmacología , Interleucina-4/química , Mutación/fisiología , Fenotipo , Fosforilación , Ingeniería de Proteínas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-4/efectos de los fármacos , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
15.
Nat Commun ; 15(1): 2071, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453915

RESUMEN

IL-11 and IL-6 activate signalling via assembly of the cell surface receptor gp130; however, it is unclear how signals are transmitted across the membrane to instruct cellular responses. Here we solve the cryoEM structure of the IL-11 receptor recognition complex to discover how differences in gp130-binding interfaces may drive signalling outcomes. We explore how mutations in the IL6ST gene encoding for gp130, which cause severe immune deficiencies in humans, impair signalling without blocking cytokine binding. We use cryoEM to solve structures of both IL-11 and IL-6 complexes with a mutant form of gp130 associated with human disease. Together with molecular dynamics simulations, we show that the disease-associated variant led to an increase in flexibility including motion within the cytokine-binding core and increased distance between extracellular domains. However, these distances are minimized as the transmembrane helix exits the membrane, suggesting a stringency in geometry for signalling and dimmer switch mode of action.


Asunto(s)
Interleucina-11 , Interleucina-6 , Humanos , Interleucina-11/genética , Interleucina-6/metabolismo , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Transducción de Señal , Receptores de Interleucina-6/genética
16.
Hum Mol Genet ; 20(24): 4840-50, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21933838

RESUMEN

Mutations in the retinitis pigmentosa GTPase regulator (RPGR) protein cause one of the most common and severe forms of inherited retinal dystrophy. In spite of numerous studies, the precise function of RPGR remains unclear, as is the mechanism by which RPGR mutations cause retinal degeneration. We have analysed the function of RPGR by RNA interference-mediated translational suppression [knockdown (KD)] using a model cellular system for studying the formation, maintenance and function of primary cilia (human telomerase-immortalized retinal pigmented epithelium 1 cells). We observed that RPGR-deficient cells exhibited reduced numbers of cilia, slower cell cycle progression and impaired attachment to fibronectin, but showed no migration defects in a wound-healing assay. RPGR KD cells showed stronger actin filaments, associated with basal dysregulation of the Akt, Erk1/2, focal adhesion kinase and Src signalling pathways, as well as a 20% reduction in ß1-integrin receptors at the cell surface and impaired fibronectin-induced signalling. Stronger actin filaments and impairment of the above signalling pathways suggest a common underlying mechanism for all of the cellular phenotypes observed in RPGR KD cells. Our data underline a novel function for RPGR in cilia formation and in the regulation of actin stress filaments, suggesting that, in the retina, it may regulate nascent photoreceptor disc formation by regulating actin-mediated membrane extension.


Asunto(s)
Actinas/metabolismo , Cilios/metabolismo , Proteínas del Ojo/metabolismo , Organogénesis , Citoesqueleto de Actina/metabolismo , Animales , Ciclo Celular , División Celular , Línea Celular , Movimiento Celular , Forma de la Célula , Células Epiteliales/citología , Células Epiteliales/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal , Adhesiones Focales/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Integrina beta1/metabolismo , Ratones , Epitelio Pigmentado de la Retina/citología , Transducción de Señal
17.
FEBS J ; 290(10): 2525-2552, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-35246947

RESUMEN

Cytokines are soluble factors vital for mammalian physiology. Cytokines elicit highly pleiotropic activities, characterized by their ability to induce a wide spectrum of functional responses in a diverse range of cell subsets, which makes their study very challenging. Cytokines activate signalling via receptor dimerization/oligomerization, triggering activation of the JAK (Janus kinase)/STAT (signal transducer and activator of transcription) signalling pathway. Given the strong crosstalk and shared usage of key components of cytokine signalling pathways, a long-standing question in the field pertains to how functional diversity is achieved by cytokines. Here, we discuss how biophysical - for example, ligand-receptor binding affinity and topology - and cellular - for example, receptor, JAK and STAT protein levels, endosomal compartment - parameters contribute to the modulation and diversification of cytokine responses. We review how these parameters ultimately converge into a common mechanism to fine-tune cytokine signalling that involves the control of the number of Tyr residues phosphorylated in the receptor intracellular domain upon cytokine stimulation. This results in different kinetics of STAT activation, and induction of specific gene expression programs, ensuring the generation of functional diversity by cytokines using a limited set of signalling intermediaries. We describe how these first principles of cytokine signalling have been exploited using protein engineering to design cytokine variants with more specific and less toxic responses for immunotherapy.


Asunto(s)
Citocinas , Transducción de Señal , Animales , Citocinas/metabolismo , Quinasas Janus/genética , Quinasas Janus/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Mamíferos/metabolismo
18.
J Immunol ; 184(10): 5678-85, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20400700

RESUMEN

TLR9 is expressed in cells of the innate immune system, as well as in B lymphocytes and their progenitors. We investigated the effect of the TLR9 ligand CpG DNA on the proliferation of pro-B cells. CpG DNA inhibits the proliferation of pro-B, but not pre-B, cells by inducing caspase-independent cell death through a pathway that requires the expression of cathepsin B. This pathway is operative in Rag-deficient mice carrying an SP6 transgene, in which B lymphopoiesis is compromised, to reduce the size of the B lymphocyte precursor compartments in the bone marrow. Thus, TLR9 signals can regulate B lymphopoiesis in vivo.


Asunto(s)
Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Catepsina B/fisiología , Diferenciación Celular/inmunología , Islas de CpG/fisiología , Células Madre/citología , Células Madre/inmunología , Animales , Subgrupos de Linfocitos B/metabolismo , Caspasas/fisiología , Catepsina B/deficiencia , Catepsina B/genética , Muerte Celular/inmunología , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Células Cultivadas , Homeostasis/inmunología , Interleucina-7/fisiología , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Células Madre/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/fisiología
19.
Sci Immunol ; 7(78): eade5686, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36459543

RESUMEN

Cytokines interact with their receptors in the extracellular space to control immune responses. How the physicochemical properties of the extracellular space influence cytokine signaling is incompletely elucidated. Here, we show that the activity of interleukin-2 (IL-2), a cytokine critical to T cell immunity, is profoundly affected by pH, limiting IL-2 signaling within the acidic environment of tumors. Generation of lactic acid by tumors limits STAT5 activation, effector differentiation, and antitumor immunity by CD8+ T cells and renders high-dose IL-2 therapy poorly effective. Directed evolution enabled selection of a pH-selective IL-2 mutein (Switch-2). Switch-2 binds the IL-2 receptor subunit IL-2Rα with higher affinity, triggers STAT5 activation, and drives CD8+ T cell effector function more potently at acidic pH than at neutral pH. Consequently, high-dose Switch-2 therapy induces potent immune activation and tumor rejection with reduced on-target toxicity in normal tissues. Last, we show that sensitivity to pH is a generalizable property of a diverse range of cytokines with broad relevance to immunity and immunotherapy in healthy and diseased tissues.


Asunto(s)
Interleucina-2 , Neoplasias , Humanos , Factor de Transcripción STAT5 , Linfocitos T CD8-positivos , Citocinas , Concentración de Iones de Hidrógeno
20.
STAR Protoc ; 2(2): 100417, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33870224

RESUMEN

Stable isotope labeling by amino acid-based high-resolution phosphoproteomics is a powerful technique that allows for direct comparison of cells stimulated under different experimental conditions. This feature makes it the ideal methodology to identify cytokine signaling networks. Here, we present an optimized protocol for the isolation and identification of phosphopeptides from IL-6-stimulated primary human Th-1 cells. For complete details on the use and execution of this protocol, please refer to Martinez-Fabregas et al. (2020).


Asunto(s)
Citocinas/metabolismo , Fosfoproteínas , Proteómica/métodos , Transducción de Señal/fisiología , Células TH1/metabolismo , Células Cultivadas , Humanos , Fosfoproteínas/análisis , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteoma/análisis , Proteoma/química , Proteoma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA