Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
FASEB J ; 37(3): e22752, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36794636

RESUMEN

Atherosclerosis is a chronic inflammatory condition of our arteries and the main underlying pathology of myocardial infarction and stroke. The pathogenesis is age-dependent, but the links between disease progression, age, and atherogenic cytokines and chemokines are incompletely understood. Here, we studied the chemokine-like inflammatory cytokine macrophage migration inhibitory factor (MIF) in atherogenic Apoe-/- mice across different stages of aging and cholesterol-rich high-fat diet (HFD). MIF promotes atherosclerosis by mediating leukocyte recruitment, lesional inflammation, and suppressing atheroprotective B cells. However, links between MIF and advanced atherosclerosis across aging have not been systematically explored. We compared effects of global Mif-gene deficiency in 30-, 42-, and 48-week-old Apoe-/- mice on HFD for 24, 36, or 42 weeks, respectively, and in 52-week-old mice on a 6-week HFD. Mif-deficient mice exhibited reduced atherosclerotic lesions in the 30/24- and 42/36-week-old groups, but atheroprotection, which in the applied Apoe-/- model was limited to lesions in the brachiocephalic artery and abdominal aorta, was not detected in the 48/42- and 52/6-week-old groups. This suggested that atheroprotection afforded by global Mif-gene deletion differs across aging stages and atherogenic diet duration. To characterize this phenotype and study the underlying mechanisms, we determined immune cells in the periphery and vascular lesions, obtained a multiplex cytokine/chemokine profile, and compared the transcriptome between the age-related phenotypes. We found that Mif deficiency promotes lesional macrophage and T-cell counts in younger but not aged mice, with subgroup analysis pointing toward a role for Trem2+ macrophages. The transcriptomic analysis identified pronounced MIF- and aging-dependent changes in pathways predominantly related to lipid synthesis and metabolism, lipid storage, and brown fat cell differentiation, as well as immunity, and atherosclerosis-relevant enriched genes such as Plin1, Ldlr, Cpne7, or Il34, hinting toward effects on lesional lipids, foamy macrophages, and immune cells. Moreover, Mif-deficient aged mice exhibited a distinct plasma cytokine/chemokine signature consistent with the notion that mediators known to drive inflamm'aging are either not downregulated or even upregulated in Mif-deficient aged mice compared with the corresponding younger ones. Lastly, Mif deficiency favored formation of lymphocyte-rich peri-adventitial leukocyte clusters. While the causative contributions of these mechanistic pillars and their interplay will be subject to future scrutiny, our study suggests that atheroprotection due to global Mif-gene deficiency in atherogenic Apoe-/- mice is reduced upon advanced aging and identifies previously unrecognized cellular and molecular targets that could explain this phenotype shift. These observations enhance our understanding of inflamm'aging and MIF pathways in atherosclerosis and may have implications for translational MIF-directed strategies.


Asunto(s)
Aterosclerosis , Factores Inhibidores de la Migración de Macrófagos , Placa Aterosclerótica , Animales , Ratones , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Aterosclerosis/metabolismo , Quimiocinas , Envejecimiento , Apolipoproteínas E/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL , Glicoproteínas de Membrana , Receptores Inmunológicos
2.
Cell Mol Life Sci ; 79(10): 512, 2022 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-36094626

RESUMEN

To fulfil its orchestration of immune cell trafficking, a network of chemokines and receptors developed that capitalizes on specificity, redundancy, and functional selectivity. The discovery of heteromeric interactions in the chemokine interactome has expanded the complexity within this network. Moreover, some inflammatory mediators, not structurally linked to classical chemokines, bind to chemokine receptors and behave as atypical chemokines (ACKs). We identified macrophage migration inhibitory factor (MIF) as an ACK that binds to chemokine receptors CXCR2 and CXCR4 to promote atherogenic leukocyte recruitment. Here, we hypothesized that chemokine-chemokine interactions extend to ACKs and that MIF forms heterocomplexes with classical chemokines. We tested this hypothesis by using an unbiased chemokine protein array. Platelet chemokine CXCL4L1 (but not its variant CXCL4 or the CXCR2/CXCR4 ligands CXCL8 or CXCL12) was identified as a candidate interactor. MIF/CXCL4L1 complexation was verified by co-immunoprecipitation, surface plasmon-resonance analysis, and microscale thermophoresis, also establishing high-affinity binding. We next determined whether heterocomplex formation modulates inflammatory/atherogenic activities of MIF. Complex formation was observed to inhibit MIF-elicited T-cell chemotaxis as assessed by transwell migration assay and in a 3D-matrix-based live cell-imaging set-up. Heterocomplexation also blocked MIF-triggered migration of microglia in cortical cultures in situ, as well as MIF-mediated monocyte adhesion on aortic endothelial cell monolayers under flow stress conditions. Of note, CXCL4L1 blocked binding of Alexa-MIF to a soluble surrogate of CXCR4 and co-incubation with CXCL4L1 attenuated MIF responses in HEK293-CXCR4 transfectants, indicating that complex formation interferes with MIF/CXCR4 pathways. Because MIF and CXCL4L1 are platelet-derived products, we finally tested their role in platelet activation. Multi-photon microscopy, FLIM-FRET, and proximity-ligation assay visualized heterocomplexes in platelet aggregates and in clinical human thrombus sections obtained from peripheral artery disease (PAD) in patients undergoing thrombectomy. Moreover, heterocomplexes inhibited MIF-stimulated thrombus formation under flow and skewed the lamellipodia phenotype of adhering platelets. Our study establishes a novel molecular interaction that adds to the complexity of the chemokine interactome and chemokine/receptor-network. MIF/CXCL4L1, or more generally, ACK/CXC-motif chemokine heterocomplexes may be target structures that can be exploited to modulate inflammation and thrombosis.


Asunto(s)
Aterosclerosis , Factores Inhibidores de la Migración de Macrófagos , Trombosis , Aterosclerosis/metabolismo , Células HEK293 , Humanos , Inflamación/metabolismo , Oxidorreductasas Intramoleculares , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factor Plaquetario 4 , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo
3.
J Biol Chem ; 295(3): 850-867, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31811089

RESUMEN

Human macrophage migration-inhibitory factor (MIF) is an evolutionarily-conserved protein that has both extracellular immune-modulating and intracellular cell-regulatory functions. MIF plays a role in various diseases, including inflammatory diseases, atherosclerosis, autoimmunity, and cancer. It serves as an inflammatory cytokine and chemokine, but also exhibits enzymatic activity. Secreted MIF binds to cell-surface immune receptors such as CD74 and CXCR4. Plants possess MIF orthologs but lack the associated receptors, suggesting functional diversification across kingdoms. Here, we characterized three MIF orthologs (termed MIF/d-dopachrome tautomerase-like proteins or MDLs) of the model plant Arabidopsis thaliana Recombinant Arabidopsis MDLs (AtMDLs) share similar secondary structure characteristics with human MIF, yet only have minimal residual tautomerase activity using either p-hydroxyphenylpyruvate or dopachrome methyl ester as substrate. Site-specific mutagenesis suggests that this is due to a distinct amino acid difference at the catalytic cavity-defining residue Asn-98. Surprisingly, AtMDLs bind to the human MIF receptors CD74 and CXCR4. Moreover, they activate CXCR4-dependent signaling in a receptor-specific yeast reporter system and in CXCR4-expressing human HEK293 transfectants. Notably, plant MDLs exert dose-dependent chemotactic activity toward human monocytes and T cells. A small molecule MIF inhibitor and an allosteric CXCR4 inhibitor counteract this function, revealing its specificity. Our results indicate cross-kingdom conservation of the receptor signaling and leukocyte recruitment capacities of human MIF by its plant orthologs. This may point toward a previously unrecognized interplay between plant proteins and the human innate immune system.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Histocompatibilidad Clase II/genética , Inmunidad Innata/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Receptores CXCR4/genética , Antígenos de Diferenciación de Linfocitos B/química , Arabidopsis/genética , Arabidopsis/inmunología , Quimiotaxis/genética , Quimiotaxis/inmunología , Secuencia Conservada/genética , Secuencia Conservada/inmunología , Citocinas/genética , Citocinas/inmunología , Células HEK293 , Antígenos de Histocompatibilidad Clase II/química , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/inmunología , Monocitos/química , Monocitos/metabolismo , Unión Proteica/genética , Receptores CXCR4/química , Homología de Secuencia , Linfocitos T/química , Linfocitos T/metabolismo
4.
Chembiochem ; 22(6): 1012-1019, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33125165

RESUMEN

Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine and atypical chemokine with a key role in inflammatory diseases including atherosclerosis. Key atherogenic functions of MIF are mediated by noncognate interaction with the chemokine receptor CXCR2. The MIF N-like loop comprising the sequence 47-56 is an important structural determinant of the MIF/CXCR2 interface and MIF(47-56) blocks atherogenic MIF activities. However, the mechanism and critical structure-activity information within this sequence have remained elusive. Here, we show that MIF(47-56) directly binds to CXCR2 to compete with MIF receptor activation. By using alanine scanning, essential and dispensable residues were identified. Moreover, MIF(cyclo10), a designed cyclized variant of MIF(47-56), inhibited key inflammatory and atherogenic MIF activities in vitro and in vivo/ex vivo, and exhibited strongly improved resistance to proteolytic degradation in human plasma in vitro, thus suggesting that it could serve as a promising basis for MIF-derived anti-atherosclerotic peptides.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/química , Péptidos Cíclicos/metabolismo , Receptores de Interleucina-8B/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular , Fluoresceínas/química , Células HEK293 , Humanos , Leucocitos/química , Leucocitos/citología , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Péptidos Cíclicos/sangre , Péptidos Cíclicos/química , Unión Proteica , Estabilidad Proteica , Receptores de Interleucina-8B/antagonistas & inhibidores , Espectrometría de Fluorescencia , Ácidos Sulfónicos/química
5.
Angew Chem Int Ed Engl ; 59(9): 3372-3384, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-31529602

RESUMEN

Protein misfolding into amyloid fibrils is linked to more than 40 as yet incurable cell- and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and type 2 diabetes. So far, however, only one of the numerous anti-amyloid molecules has reached patients. This Minireview gives an overview of molecular strategies and peptide chemistry "tools" to design, develop, and discover peptide-based molecules as anti-amyloid drug candidates. We focus on two major inhibitor rational design strategies: 1) the oldest and most common strategy, based on molecular recognition elements of amyloid self-assembly, and 2) a more recent approach, based on cross-amyloid interactions. We discuss why peptide-based amyloid inhibitors, in particular their advanced generations, can be promising leads or candidates for anti-amyloid drugs as well as valuable tools for deciphering amyloid-mediated cell damage and its link to disease pathogenesis.


Asunto(s)
Amiloide/antagonistas & inhibidores , Péptidos/química , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diseño de Fármacos , Humanos , Péptidos/metabolismo , Péptidos/farmacología , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína
6.
Angew Chem Int Ed Engl ; 59(31): 12837-12841, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-31999880

RESUMEN

Amyloid self-assembly of islet amyloid polypeptide (IAPP) is linked to pancreatic inflammation, ß-cell degeneration, and the pathogenesis of type 2 diabetes (T2D). The multifunctional host-defence peptides (HDPs) cathelicidins play crucial roles in inflammation. Here, we show that the antimicrobial and immunomodulatory polypeptide human cathelicidin LL-37 binds IAPP with nanomolar affinity and effectively suppresses its amyloid self-assembly and related pancreatic ß-cell damage in vitro. In addition, we identify key LL-37 segments that mediate its interaction with IAPP. Our results suggest a possible protective role for LL-37 in T2D pathogenesis and offer a molecular basis for the design of LL-37-derived peptides that combine antimicrobial, immunomodulatory, and T2D-related anti-amyloid functions as promising candidates for multifunctional drugs.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Multimerización de Proteína/efectos de los fármacos , Secuencia de Aminoácidos , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Catelicidinas
7.
Angew Chem Int Ed Engl ; 59(14): 5771-5781, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-31863711

RESUMEN

Designed peptides derived from the islet amyloid polypeptide (IAPP) cross-amyloid interaction surface with Aß (termed interaction surface mimics or ISMs) have been shown to be highly potent inhibitors of Aß amyloid self-assembly. However, the molecular mechanism of their function is not well understood. Using solution-state and solid-state NMR spectroscopy in combination with ensemble-averaged dynamics simulations and other biophysical methods including TEM, fluorescence spectroscopy and microscopy, and DLS, we characterize ISM structural preferences and interactions. We find that the ISM peptide R3-GI is highly dynamic, can adopt a ß-like structure, and oligomerizes into colloid-like assemblies in a process that is reminiscent of liquid-liquid phase separation (LLPS). Our results suggest that such assemblies yield multivalent surfaces for interactions with Aß40. Sequestration of substrates into these colloid-like structures provides a mechanistic basis for ISM function and the design of novel potent anti-amyloid molecules.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/química , Fragmentos de Péptidos/antagonistas & inhibidores , Péptidos/química , Secuencia de Aminoácidos , Péptidos beta-Amiloides/metabolismo , Microscopía Fluorescente , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Especificidad por Sustrato
8.
FASEB J ; 32(8): 4428-4443, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29543531

RESUMEN

The inflammatory cytokine macrophage migration-inhibitory factor (MIF) promotes atherosclerosis via lesional monocyte and T-cell recruitment. B cells have emerged as important components in atherogenesis, but the interaction between MIF and B cells in atherogenesis is unknown. Here, we investigated the atherosclerotic phenotype of Mif-gene deletion in Apoe-/- mice. Apoe-/- Mif-/- mice fed a Western diet exhibited strongly reduced atherosclerotic lesions in brachiocephalic artery (BC) and abdominal aorta compared with controls. This phenotype was accompanied by reduced circulating B cells. Flow cytometry revealed a B-cell developmental defect with increased premature and immature B-cell counts in bone marrow (BM) of Apoe-/- Mif-/- mice and diminished B-cell numbers in spleen. This finding was linked with a decreased expression of Baff-R and differentiation-driving transcription factors at the immature B-cell stage, whereas peritoneal B cells exhibited unchanged CD80 and CD86 expression but vastly decreased CD9 and elevated CD23 levels, indicating that the developmental block favors the generation of immature, egressing, and reactive B cells. Mif deficiency did not affect absolute B-cell numbers in the vessel wall but favored a relative increase of B cells in the atheroprone BC region and the appearance of periadventitial B-cell-rich clusters. Of note, Mif-/- mice exhibited a significant increase in oxidized low-density lipoprotein (oxLDL)-specific antibodies after the injection of oxLDL, indicating that Mif deficiency is associated with higher sensitivity of B cells against natural-occurring antigens such as oxLDL. Importantly, Apoe-/- mice adoptively transplanted with Apoe-/-Mif-/- BM showed reduced peripheral B cells compared with Apoe-/- BM transplantation but no atheroprotection in the BC; also, whereas there was a selective increase in atheroprotective IgM-anti-oxLDL-antibodies in global Mif deficiency, BM-specific Mif deficiency also led to elevated proatherogenic anti-oxLDL-IgG. Together, these findings reveal a novel link between MIF and B cells in atherogenesis. Protection from atherosclerosis by Mif deficiency is associated with enhanced B-cell hypersensitivity, which in global but not BM-restricted Mif deficiency favors an atheroprotective autoantibody profile in atherosclerotic mice. Targeting MIF may induce protective B-cell responses in atherosclerosis.-Schmitz, C., Noels, H., El Bounkari, O., Straussfeld, E., Megens, R. T. A., Sternkopf, M., Alampour-Rajabi, S., Krammer, C., Tilstam, P. V., Gerdes, N., Bürger, C., Kapurniotu, A., Bucala, R., Jankowski, J., Weber, C., Bernhagen, J. Mif-deficiency favors an atheroprotective autoantibody phenotype in atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Autoanticuerpos/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Animales , Aorta/metabolismo , Apolipoproteínas E/metabolismo , Linfocitos B/metabolismo , Diferenciación Celular/fisiología , Femenino , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Fenotipo , Placa Aterosclerótica/metabolismo
9.
J Biol Chem ; 292(35): 14587-14602, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28684415

RESUMEN

The interaction of the intrinsically disordered polypeptide islet amyloid polypeptide (IAPP), which is associated with type 2 diabetes (T2D), with the Alzheimer's disease amyloid-ß (Aß) peptide modulates their self-assembly into amyloid fibrils and may link the pathogeneses of these two cell-degenerative diseases. However, the molecular determinants of this interaction remain elusive. Using a systematic alanine scan approach, fluorescence spectroscopy, and other biophysical methods, including heterocomplex pulldown assays, far-UV CD spectroscopy, the thioflavin T binding assay, transmission EM, and molecular dynamics simulations, here we identified single aromatic/hydrophobic residues within the amyloid core IAPP region as hot spots or key residues of its cross-interaction with Aß40(42) peptide. Importantly, we also find that none of these residues in isolation plays a key role in IAPP self-assembly, whereas simultaneous substitution of four aromatic/hydrophobic residues with Ala dramatically impairs both IAPP self-assembly and hetero-assembly with Aß40(42). Furthermore, our experiments yielded several novel IAPP analogs, whose sequences are highly similar to that of IAPP but have distinct amyloid self- or cross-interaction potentials. The identified similarities and major differences controlling IAPP cross-peptide interaction with Aß40(42) versus its amyloid self-assembly offer a molecular basis for understanding the underlying mechanisms. We propose that these insights will aid in designing intervention strategies and novel IAPP analogs for the management of type 2 diabetes, Alzheimer's disease, or other diseases related to IAPP dysfunction or cross-amyloid interactions.


Asunto(s)
Aminoácidos/metabolismo , Péptidos beta-Amiloides/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Modelos Moleculares , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Sustitución de Aminoácidos , Aminoácidos/química , Aminoácidos Aromáticos , Péptidos beta-Amiloides/síntesis química , Péptidos beta-Amiloides/química , Dicroismo Circular , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Polipéptido Amiloide de los Islotes Pancreáticos/síntesis química , Polipéptido Amiloide de los Islotes Pancreáticos/química , Cinética , Metilación , Microscopía Electrónica de Transmisión , Simulación de Dinámica Molecular , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Estabilidad Proteica , Estructura Secundaria de Proteína , Técnicas de Síntesis en Fase Sólida , Solubilidad , Espectrometría de Fluorescencia
10.
Angew Chem Int Ed Engl ; 57(44): 14503-14508, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29908130

RESUMEN

Amyloid self-assembly is linked to the pathogenesis of Alzheimer's disease (AD) and type 2 diabetes (T2D), but so far, no anti-amyloid compound has reached the clinic. Macrocyclic peptides belong to the most attractive drug candidates. Herein we present macrocyclic peptides (MCIPs) designed using minimal IAPP-derived recognition elements as a novel class of nanomolar amyloid inhibitors of both Aß40(42) and IAPP or Aß40(42) alone and show that chirality controls inhibitor selectivity. Sequence optimization led to the discovery of an Aß40(42)-selective MCIP exhibiting high proteolytic stability in human plasma and human blood-brain barrier (BBB) crossing ability in a cell model, two highly desirable properties for anti-amyloid AD drugs. Owing to their favorable properties, MCIPs should serve as leads for macrocyclic peptide-based anti-amyloid drugs and scaffolds for the design of small-molecule peptidomimetics for targeting amyloidogenesis in AD or in both AD and T2D.


Asunto(s)
Amiloide/antagonistas & inhibidores , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Nanotecnología , Péptidos/química , Péptidos/farmacología , Secuencia de Aminoácidos
11.
J Biol Chem ; 291(30): 15881-95, 2016 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-27226569

RESUMEN

An emerging number of non-chemokine mediators are found to bind to classical chemokine receptors and to elicit critical biological responses. Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine that exhibits chemokine-like activities through non-cognate interactions with the chemokine receptors CXCR2 and CXCR4, in addition to activating the type II receptor CD74. Activation of the MIF-CXCR2 and -CXCR4 axes promotes leukocyte recruitment, mediating the exacerbating role of MIF in atherosclerosis and contributing to the wealth of other MIF biological activities. Although the structural basis of the MIF-CXCR2 interaction has been well studied and was found to engage a pseudo-ELR and an N-like loop motif, nothing is known about the regions of CXCR4 and MIF that are involved in binding to each other. Using a genetic strain of Saccharomyces cerevisiae that expresses a functional CXCR4 receptor, site-specific mutagenesis, hybrid CXCR3/CXCR4 receptors, pharmacological reagents, peptide array analysis, chemotaxis, fluorescence spectroscopy, and circular dichroism, we provide novel molecular information about the structural elements that govern the interaction between MIF and CXCR4. The data identify similarities with classical chemokine-receptor interactions but also provide evidence for a partial allosteric agonist compared with CXCL12 that is possible due to the two binding sites of CXCR4.


Asunto(s)
Quimiocina CXCL12 , Oxidorreductasas Intramoleculares , Factores Inhibidores de la Migración de Macrófagos , Receptores CXCR4 , Regulación Alostérica , Animales , Células CHO , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Cricetinae , Cricetulus , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptores CXCR4/química , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Angew Chem Int Ed Engl ; 54(44): 13095-100, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26336938

RESUMEN

The design of inhibitors of protein-protein interactions mediating amyloid self-assembly is a major challenge mainly due to the dynamic nature of the involved structures and interfaces. Interactions of amyloidogenic polypeptides with other proteins are important modulators of self-assembly. Here we present a hot-segment-linking approach to design a series of mimics of the IAPP cross-amyloid interaction surface with Aß (ISMs) as nanomolar inhibitors of amyloidogenesis and cytotoxicity of Aß, IAPP, or both polypeptides. The nature of the linker determines ISM structure and inhibitory function including both potency and target selectivity. Importantly, ISMs effectively suppress both self- and cross-seeded IAPP self-assembly. Our results provide a novel class of highly potent peptide leads for targeting protein aggregation in Alzheimer's disease, type 2 diabetes, or both diseases and a chemical approach to inhibit amyloid self-assembly and pathogenic interactions of other proteins as well.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Diseño de Fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Agregado de Proteínas/efectos de los fármacos , Propiedades de Superficie
13.
J Am Chem Soc ; 135(4): 1317-29, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23265086

RESUMEN

The etiology of Alzheimer's disease depends on the relative abundance of different amyloid-ß (Aß) peptide species. These peptides are produced by sequential proteolytic cleavage within the transmembrane helix of the 99 residue C-terminal fragment of the amyloid precursor protein (C99) by the intramembrane protease γ-secretase. Intramembrane proteolysis is thought to require local unfolding of the substrate helix, which has been proposed to be cleaved as a homodimer. Here, we investigated the backbone dynamics of the substrate helix. Amide exchange experiments of monomeric recombinant C99 and of synthetic transmembrane domain peptides reveal that the N-terminal Gly-rich homodimerization domain exchanges much faster than the C-terminal cleavage region. MD simulations corroborate the differential backbone dynamics, indicate a bending motion at a diglycine motif connecting dimerization and cleavage regions, and detect significantly different H-bond stabilities at the initial cleavage sites. Our results are consistent with the following hypotheses about cleavage of the substrate: First, the GlyGly hinge may precisely position the substrate within γ-secretase such that its catalytic center must start proteolysis at the known initial cleavage sites. Second, the ratio of cleavage products formed by subsequent sequential proteolysis could be influenced by differential extents of solvation and by the stabilities of H-bonds at alternate initial sites. Third, the flexibility of the Gly-rich domain may facilitate substrate movement within the enzyme during sequential proteolysis. Fourth, dimerization may affect substrate processing by decreasing the dynamics of the dimerization region and by increasing that of the C-terminal part of the cleavage region.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Termodinámica , Secretasas de la Proteína Precursora del Amiloide/química , Péptidos beta-Amiloides/síntesis química , Péptidos beta-Amiloides/química , Modelos Moleculares
14.
Sci Rep ; 13(1): 11611, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37464010

RESUMEN

Non-melanoma skin cancer (NMSC) is the most common cancer in Caucasians worldwide. We investigated the pathophysiological role of MIF and its homolog D-DT in UVB- and chemically induced NMSC using Mif-/-, D-dt-/- and Mif-/-/D-dt-/- mice on a hairless SKH1 background. Knockout of both cytokines showed similar attenuating effects on inflammation after acute UVB irradiation and tumor formation during chronic UVB irradiation, without additive protective effects noted in double knockout mice, indicating that both cytokines activate a similar signaling threshold. In contrast, genetic deletion of Mif and D-dt had no major effects on chemically induced skin tumors. To get insight into the contributing mechanisms, we used an in vitro 3D skin model with incorporated macrophages. Application of recombinant MIF and D-DT led to an accumulation of macrophages within the epidermal part that could be reversed by selective inhibitors of MIF and D-DT pathways. In summary, our data indicate that MIF and D-DT contribute to the development and progression of UVB- but not chemically induced NMSC, a role at least partially accounted by effects of both cytokines on epidermal macrophage accumulation. These data highlight that MIF and D-DT are both potential therapeutic targets for the prevention of photocarcinogenesis but not chemical carcinogenesis.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos , Neoplasias Cutáneas , Animales , Ratones , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones Noqueados , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética
15.
FASEB J ; 25(3): 894-906, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21106938

RESUMEN

Macrophage migration inhibitory factor (MIF) is a cytokine that mediates inflammatory diseases. MIF promotes atherogenic leukocyte recruitment through a promiscuous, yet highly affine, interaction with CXCR2 and CXCR4. Binding to CXCR2 is dependent on a pseudo-(E)LR motif in MIF, but a second interaction site has been elusive. Here we identified an N-like loop in MIF, suggesting that MIF binding to CXCR2 follows the 2-site binding mode of bona fide chemokines. For MIF, the model predicts interactions between the N-like loop and the CXCR2 N domain (site 1) and pseudo-(E)LR and extracellular loops (ELs) of CXCR2 (site 2). Applying biophysical and peptide array analysis, we demonstrated an interaction between MIF and the CXCR2 N domain, which was pseudo-(E)LR independent. Peptide array analysis also indicated that the pseudo-(E)LR motif is responsible for MIF binding to EL2 and 3. Notably, peptides MIF-(40-49) and MIF-(47-56), representing N-like-loop-derived peptides, but not a scrambled control peptide, significantly blocked MIF/CXCR2 binding, MIF-mediated monocyte arrest under flow on aortic endothelial cells in vitro (IC(50): 1.24×10(-6) M), and MIF-dependent monocyte adhesion to atherosclerotic mouse carotid arteries in vivo. Thus, the N-like loop in MIF is critical for MIF's noncognate interaction with CXCR2 and proatherogenic functions. The 2-site binding model that explains chemokine receptor activation also applies to MIF.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/fisiología , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/metabolismo , Animales , Aorta/citología , Apolipoproteínas E/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Sitios de Unión/fisiología , Unión Competitiva/fisiología , Dicroismo Circular , Células Endoteliales/citología , Células HEK293 , Humanos , Interleucina-8/metabolismo , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Monocitos/citología , Unión Proteica/fisiología , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores de Interleucina-8B/genética , Relación Estructura-Actividad
16.
Nat Commun ; 13(1): 5004, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36008417

RESUMEN

Amyloid self-assembly is linked to numerous devastating cell-degenerative diseases. However, designing inhibitors of this pathogenic process remains a major challenge. Cross-interactions between amyloid-ß peptide (Aß) and islet amyloid polypeptide (IAPP), key polypeptides of Alzheimer's disease (AD) and type 2 diabetes (T2D), have been suggested to link AD with T2D pathogenesis. Here, we show that constrained peptides designed to mimic the Aß amyloid core (ACMs) are nanomolar cross-amyloid inhibitors of both IAPP and Aß42 and effectively suppress reciprocal cross-seeding. Remarkably, ACMs act by co-assembling with IAPP or Aß42 into amyloid fibril-resembling but non-toxic nanofibers and their highly ordered superstructures. Co-assembled nanofibers exhibit various potentially beneficial features including thermolability, proteolytic degradability, and effective cellular clearance which are reminiscent of labile/reversible functional amyloids. ACMs are thus promising leads for potent anti-amyloid drugs in both T2D and AD while the supramolecular nanofiber co-assemblies should inform the design of novel functional (hetero-)amyloid-based nanomaterials for biomedical/biotechnological applications.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Diabetes Mellitus Tipo 2 , Nanofibras , Enfermedad de Alzheimer/tratamiento farmacológico , Amiloide/farmacología , Péptidos beta-Amiloides/química , Proteínas Amiloidogénicas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química
17.
Biochemistry ; 50(38): 8202-12, 2011 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-21848289

RESUMEN

Hairpin peptides bearing cross-strand Trp-Trp and Tyr-Tyr pairs at non-H-bonded strand sites modulate the aggregation of two unrelated amyloidogenic systems, human pancreatic amylin (hAM) and α-synuclein (α-syn), associated with type II diabetes and Parkinson's disease, respectively. In the case of hAM, we have previously reported that inhibition of amyloidogenesis is observed as an increase in the lag time to amyloid formation and a diminished thioflavin (ThT) fluorescence response. In this study, a reduced level of hAM fibril formation is confirmed by transmission electron microscopy imaging. Several of the hairpins tested were significantly more effective inhibitors than rat amylin. Moreover, a marked inhibitory effect on hAM-associated cytotoxicity by the more potent hairpin peptide is demonstrated. In the case of α-syn, the dominant effect of active hairpins was, besides a weakened ThT fluorescence response, the earlier appearance of insoluble aggregates that do not display amyloid characteristics with the few fibrils observed having abnormal morphology. We attribute the alteration of the α-synuclein aggregation pathway observed to the capture of a preamyloid state and diversion to nonamyloidogenic aggregates. These ß-hairpins represent a new class of amyloid inhibitors that bear no sequence similarity to the amyloid-producing polypeptides that are inhibited. A mechanistic rationale for these effects is proposed.


Asunto(s)
Amiloide/antagonistas & inhibidores , Amiloide/biosíntesis , Péptidos/química , Péptidos/farmacología , Secuencia de Aminoácidos , Amiloide/química , Animales , Benzotiazoles , Diseño de Fármacos , Humanos , Técnicas In Vitro , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/ultraestructura , Microscopía Electrónica de Transmisión , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/genética , Pliegue de Proteína , Multimerización de Proteína/efectos de los fármacos , Ratas , Proteínas Recombinantes/farmacología , Tiazoles/química , alfa-Sinucleína/química , alfa-Sinucleína/farmacología
18.
Chembiochem ; 12(9): 1313-22, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21630409

RESUMEN

Alzheimer's disease (AD) and type 2 diabetes (T2D) are linked to the self-association of ß-amyloid peptide (Aß) and islet amyloid polypeptide (IAPP), respectively. We have shown that IAPP-GI, a soluble IAPP analogue and mimic of nonamyloidogenic and nontoxic IAPP, binds Aß with high affinity and blocks its cytotoxic self-assembly and fibrillogenesis. We have also shown that IAPP and Aß interact with each other into nonfibrillar and nontoxic heterocomplexes that suppress cytotoxic self-association by both polypeptides. The Aß-IAPP interaction might thus be a molecular link between AD and T2D. We studied the role of individual IAPP-GI and IAPP regions in their inhibitory function on Aß40 self-association and cytotoxicity. We found that the presence of the two hot-spot regions of the Aß-IAPP interaction interface in IAPP(8-28) is not sufficient for inhibitory function and that, in addition to IAPP(8-28), the presence of the N-terminal region IAPP(1-7) is absolutely required. By contrast, the C-terminal region, IAPP(30-37), is not required although its presence together with IAPP(1-7) in IAPP-GI results in a marked enhancement of the inhibitory effect as compared to IAPP(1-28)-GI. We suggest that the inhibitory effect of IAPP-GI and IAPP on Aß40 fibrillogenesis and cell toxicity is mediated primarily by interactions involving the hot regions of the Aß-IAPP interaction interface and the N terminus of IAPP while a concerted and likely structure-stabilizing action of the N- and C-terminal IAPP regions potentiates this effect. These results identify important molecular determinants of the amyloid suppressing function of the Aß40-IAPP interaction and could contribute to the design of novel inhibitors of Aß40 aggregation and cell degeneration.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Amiloide , Polipéptido Amiloide de los Islotes Pancreáticos , Oligopéptidos/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Secuencia de Aminoácidos , Amiloide/genética , Amiloide/farmacología , Dicroismo Circular , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Polipéptido Amiloide de los Islotes Pancreáticos/toxicidad , Datos de Secuencia Molecular , Oligopéptidos/genética , Unión Proteica , Espectrometría de Fluorescencia
19.
Proc Natl Acad Sci U S A ; 105(42): 16278-83, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18852457

RESUMEN

We have recently identified the archaic cytokine macrophage migration inhibitory factor (MIF) as a non-canonical ligand of the CXC chemokine receptors CXCR2 and CXCR4 in inflammatory and atherogenic cell recruitment. Because its affinity for CXCR2 was particularly high, we hypothesized that MIF may feature structural motives shared by canonical CXCR2 ligands, namely the conserved N-terminal Glu-Leu-Arg (ELR) motif. Sequence alignment and structural modeling indeed revealed a pseudo-(E)LR motif (Asp-44-X-Arg-11) constituted by non-adjacent residues in neighboring loops but with identical parallel spacing as in the authentic ELR motif. Structure-function analysis demonstrated that mutation of residues R11, D44, or both preserve proper folding and the intrinsic catalytic property of MIF but severely compromises its binding to CXCR2 and abrogates MIF/CXCR2-mediated functions in chemotaxis and arrest of monocytes on endothelium under flow conditions. R11A-MIF and the R11A/D44A-MIF double-mutant exhibited a pronounced defect in triggering leukocyte recruitment to early atherosclerotic endothelium in carotid arteries perfused ex vivo and upon application in a peritonitis model. The function of D44A-MIF in peritoneal leukocyte recruitment was preserved as a result of compensatory use of CXCR4. In conjunction, our data identify a pseudo-(E)LR motif as the structural determinant for MIF's activity as a non-canonical CXCR2 ligand, epitomizing the structural resemblance of chemokine-like ligands with chemokines and enabling selective targeting of pro-inflammatory MIF/CXCR2 interactions.


Asunto(s)
Aterosclerosis/metabolismo , Leucocitos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/metabolismo , Animales , Aterosclerosis/genética , Movimiento Celular , Células Cultivadas , Humanos , Inflamación/genética , Inflamación/metabolismo , Leucocitos/citología , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Noqueados , Modelos Moleculares , Mutación/genética , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Receptores de Interleucina-8B/genética
20.
Nat Commun ; 11(1): 5981, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239628

RESUMEN

Targeting a specific chemokine/receptor axis in atherosclerosis remains challenging. Soluble receptor-based strategies are not established for chemokine receptors due to their discontinuous architecture. Macrophage migration-inhibitory factor (MIF) is an atypical chemokine that promotes atherosclerosis through CXC-motif chemokine receptor-4 (CXCR4). However, CXCR4/CXCL12 interactions also mediate atheroprotection. Here, we show that constrained 31-residue-peptides ('msR4Ms') designed to mimic the CXCR4-binding site to MIF, selectively bind MIF with nanomolar affinity and block MIF/CXCR4 without affecting CXCL12/CXCR4. We identify msR4M-L1, which blocks MIF- but not CXCL12-elicited CXCR4 vascular cell activities. Its potency compares well with established MIF inhibitors, whereas msR4M-L1 does not interfere with cardioprotective MIF/CD74 signaling. In vivo-administered msR4M-L1 enriches in atherosclerotic plaques, blocks arterial leukocyte adhesion, and inhibits atherosclerosis and inflammation in hyperlipidemic Apoe-/- mice in vivo. Finally, msR4M-L1 binds to MIF in plaques from human carotid-endarterectomy specimens. Together, we establish an engineered GPCR-ectodomain-based mimicry principle that differentiates between disease-exacerbating and -protective pathways and chemokine-selectively interferes with atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Receptores CXCR4/metabolismo , Anciano , Animales , Antígenos CD/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/cirugía , Sitios de Unión , Arteria Carótida Común/patología , Arteria Carótida Común/cirugía , Quimiocina CXCL12/metabolismo , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Endarterectomía Carotidea , Femenino , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados para ApoE , Persona de Mediana Edad , Fragmentos de Péptidos/uso terapéutico , Receptores CXCR4/química , Receptores CXCR4/ultraestructura , Sialiltransferasas/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA