Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
FEBS J ; 291(19): 4372-4391, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39003565

RESUMEN

Tumor necrosis factor (TNF) is a pro-inflammatory cytokine and its functional homotrimeric form interacts with the TNF receptor (TNFR) to activate downstream apoptotic, necroptotic, and inflammatory signaling pathways. Excessive activation of these pathways leads to various inflammatory diseases, which makes TNF a promising therapeutic target. Here, 12-mer peptides were selected from the interface of TNF-TNFR based upon their relative binding energies and were named 'TNF-inhibiting decoys' (TIDs). These decoy peptides inhibited TNF-mediated secretion of cytokines and cell death, as well as activation of downstream signaling effectors. Effective TIDs inhibited TNF signaling by disrupting the formation of TNF's functional homotrimeric form. Among derivatives of TIDs, TID3c showed slightly better efficacy in cell-based assays by disrupting TNF trimer formation. Moreover, TID3c oligomerized TNF to a high molecular weight configuration. In silico modeling and simulations revealed that TID3c and its parent peptide, TID3, form a stable complex with TNF through hydrogen bonds and electrostatic interactions, which makes them the promising lead to develop peptide-based anti-TNF therapeutics.


Asunto(s)
Péptidos , Transducción de Señal , Factor de Necrosis Tumoral alfa , Humanos , Transducción de Señal/efectos de los fármacos , Péptidos/farmacología , Péptidos/química , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores del Factor de Necrosis Tumoral/química , Multimerización de Proteína/efectos de los fármacos , Unión Proteica , Apoptosis/efectos de los fármacos , Simulación de Dinámica Molecular
2.
J Chem Inf Model ; 64(13): 5090-5107, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38904299

RESUMEN

The aberrant secretion of proinflammatory cytokines by immune cells is the principal cause of inflammatory diseases, such as systemic lupus erythematosus and rheumatoid arthritis. Toll-like receptor 7 (TLR7) and TLR9, sequestered to the endosomal compartment of dendritic cells and macrophages, are closely associated with the initiation and progression of these diseases. Therefore, the development of drugs targeting dysregulated endosomal TLRs is imperative to mitigate systemic inflammation. Here, we applied the principles of computer-aided drug discovery to identify a novel low-molecular-weight compound, TLR inhibitory compound 10 (TIC10), and its potent derivative (TIC10g), which demonstrated dual inhibition of TLR7 and TLR9 signaling pathways. Compared to TIC10, TIC10g exhibited a more pronounced inhibition of the TLR7- and TLR9-mediated secretion of the proinflammatory cytokine tumor necrosis factor-α in a mouse macrophage cell line and mouse bone marrow dendritic cells in a concentration-dependent manner. While TIC10g slightly prevented TLR3 and TLR8 activation, it had no impact on cell surface TLRs (TLR1/2, TLR2/6, TLR4, or TLR5), indicating its selectivity for TLR7 and TLR9. Additionally, mechanistic studies suggested that TIC10g interfered with TLR9 activation by CpG DNA and suppressed downstream pathways by directly binding to TLR9. Western blot analysis revealed that TIC10g downregulated the phosphorylation of the p65 subunit of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinases (MAPKs), including extracellular-signal-regulated kinase, p38-MAPK, and c-Jun N-terminal kinase. These findings indicate that the novel ligand, TIC10g, is a specific dual inhibitor of endosomal TLRs (TLR7 and TLR9), disrupting MAPK- and NF-κB-mediated proinflammatory gene expression.


Asunto(s)
Bibliotecas de Moléculas Pequeñas , Receptor Toll-Like 7 , Receptor Toll-Like 9 , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 7/metabolismo , Animales , Ratones , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Descubrimiento de Drogas , Simulación del Acoplamiento Molecular , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Humanos
3.
Sci Signal ; 15(759): eabi8713, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36346838

RESUMEN

Excessive signaling by the proinflammatory cytokine TNF is involved in several autoimmune diseases, including rheumatoid arthritis (RA). However, unlike the approved biologics currently used to treat this and other conditions, commercially available small-molecule inhibitors of TNF trimerization are cytotoxic or exhibit low potency. Here, we report a TNF-inhibitory molecule (TIM) that reduced TNF signaling in vitro and was an effective treatment in a mouse model of RA. The initial lead compound, TIM1, attenuated TNF-induced apoptosis of human and mouse cells by delaying the induction of proinflammatory NF-κB and MAPK signaling and caspase 3- and caspase 8-dependent apoptosis. TIM1 inhibited the secretion of the proinflammatory cytokines IL-6 and IL-8 by disrupting TNF homotrimerization, thereby preventing its association with the TNF receptor. In a mouse model of collagen-induced polyarthritis, the more potent TIM1 analog TIM1c was orally bioavailable and reduced paw swelling, histological indicators of knee joint pathology, inflammatory infiltration of the joint, and the overall arthritis index. Orally delivered TIM1c showed immunological effects similar to those elicited by intraperitoneal injection of the FDA-approved TNF receptor decoy etanercept. Thus, TIM1c is a promising lead compound for the development of small-molecule therapies for the treatment of RA and other TNF-dependent systemic inflammation disorders.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Ratones , Humanos , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico , Inhibidores del Factor de Necrosis Tumoral , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/tratamiento farmacológico , FN-kappa B , Citocinas , Receptores del Factor de Necrosis Tumoral , Modelos Animales de Enfermedad
4.
J Mol Biol ; 434(11): 167604, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35662468

RESUMEN

Cell-penetrating peptides (CPPs) translocate into the cell as various biologically active conjugates and possess numerous biomedical applications. Several machine learning-based predictors have been proposed in the past, but they mostly focus on identifying only CPPs. We proposed a two-layered predictor in 2018 in order to predict CPPs and their uptake efficiency simultaneously. While MLCPP has gained widespread access to research, further improvements are needed to enhance its practical application. A new version of MLCPP is presented in this study called MLCPP 2.0, an interpretable stacking model that identifies CPPs and their strength of uptake efficiency. We updated the benchmarking dataset, explored 17 different sequence-based feature encoding algorithms, and used seven different conventional machine learning classifiers. With multiple 10-fold cross-validation, we constructed 119 baseline models whose predicted probability values were merged and treated as a new feature vector. In a systematic way, a feature set and a classifier are identified that are optimal for predicting the CPP and uptake efficiency separately. The MLCPP 2.0 model achieved outstanding performance on the independent test set, significantly outperforming the existing state-of-the-art predictors. Hence, we expect that our proposed MLCPP 2.0 will facilitate the design of hypothesis-driven experiments by enabling the discovery of novel CPPs. MLCPP 2.0 is freely accessible at https://balalab-skku.org/mlcpp2/.


Asunto(s)
Péptidos de Penetración Celular , Uso de Internet , Aprendizaje Automático , Programas Informáticos , Algoritmos , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/metabolismo , Biología Computacional , Transporte de Proteínas
5.
Int J Mol Sci ; 21(24)2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33322533

RESUMEN

Direct inhibition of tumor necrosis factor-alpha (TNF-α) action is considered a promising way to prevent or treat TNF-α-associated diseases. The trimeric form of TNF-α binds to its receptor (TNFR) and activates the downstream signaling pathway. The interaction of TNF-α with molecular-grade dimethyl sulfoxide (DMSO) in an equal volumetric ratio renders TNF-α inert, in this state, TNF-α fails to activate TNFR. Here, we aimed to examine the inhibition of TNF-α function by various concentrations of DMSO. Its higher concentration led to stronger attenuation of TNF-α-induced cytokine secretion by fibroblasts, and of their death. We found that this inhibition was mediated by a perturbation in the formation of the functional TNF-α trimer. Molecular dynamics simulations revealed a transient interaction between DMSO molecules and the central hydrophobic cavity of the TNF-α homodimer, indicating that a brief interaction of DMSO with the TNF-α homodimer may disrupt the formation of the functional homotrimer. We also found that the sensitizing effect of actinomycin D on TNF-α-induced cell death depends upon the timing of these treatments and on the cell type. This study will help to select an appropriate concentration of DMSO as a working solvent for the screening of water-insoluble TNF-α inhibitors.


Asunto(s)
Dimetilsulfóxido/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Western Blotting , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Cells ; 9(7)2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32660060

RESUMEN

Toll-like receptors (TLRs) play a fundamental role in the inflammatory response against invading pathogens. However, the dysregulation of TLR-signaling pathways is implicated in several autoimmune/inflammatory diseases. Here, we show that a novel small molecule TLR-inhibitor (TAC5) and its derivatives TAC5-a, TAC5-c, TAC5-d, and TAC5-e predominantly antagonized poly(I:C) (TLR3)-, imiquimod (TLR7)-, TL8-506 (TLR8)-, and CpG-oligodeoxynucleotide (TLR9)-induced signaling pathways. TAC5 and TAC5-a significantly hindered the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), reduced the phosphorylation of mitogen-activated protein kinases, and inhibited the secretion of tumor necrosis factor-α (TNF-α) and interleukin-6. Besides, TAC5-a prevented the progression of psoriasis and systemic lupus erythematosus (SLE) in mice. Interestingly, TAC5 and TAC5-a did not affect Pam3CSK4 (TLR1/2)-, FSL-1 (TLR2/6)-, or lipopolysaccharide (TLR4)-induced TNF-α secretion, indicating their specificity towards endosomal TLRs (TLR3/7/8/9). Collectively, our data suggest that the TAC5 series of compounds are potential candidates for treating autoimmune diseases such as psoriasis or SLE.


Asunto(s)
Antiinflamatorios/farmacología , Factores Inmunológicos/farmacología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Receptores Toll-Like/antagonistas & inhibidores , Animales , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Sitios de Unión , Endosomas/metabolismo , Femenino , Factores Inmunológicos/química , Factores Inmunológicos/uso terapéutico , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Unión Proteica , Relación Estructura-Actividad Cuantitativa , Células RAW 264.7 , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Receptores Toll-Like/química , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Int J Mol Sci ; 21(8)2020 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-32325904

RESUMEN

Toll-like receptor 3 (TLR3) provides the host with antiviral defense by initiating an immune signaling cascade for the production of type I interferons. The X-ray structures of isolated TLR3 ectodomain (ECD) and transmembrane (TM) domains have been reported; however, the structure of a membrane-solvated, full-length receptor remains elusive. We investigated an all-residue TLR3 model embedded inside a phospholipid bilayer using molecular dynamics simulations. The TLR3-ECD exhibited a ~30°-35° tilt on the membrane due to the electrostatic interaction between the N-terminal subdomain and phospholipid headgroups. Although the movement of dsRNA did not affect the dimer integrity of TLR3, its sugar-phosphate backbone was slightly distorted with the orientation of the ECD. TM helices exhibited a noticeable tilt and curvature but maintained a consistent crossing angle, avoiding the hydrophobic mismatch with the bilayer. Residues from the αD helix and the CD and DE loops of the Toll/interleukin-1 receptor (TIR) domains were partially absorbed into the lower leaflet of the bilayer. We found that the previously unknown TLR3-TIR dimerization interface could be stabilized by the reciprocal contact between αC and αD helices of one subunit and the αC helix and the BB loop of the other. Overall, the present study can be helpful to understand the signaling-competent form of TLR3 in physiological environments.


Asunto(s)
Membrana Dobles de Lípidos/química , Fosfolípidos/química , Receptor Toll-Like 3/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Fosfolípidos/metabolismo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Bicatenario/química , ARN Bicatenario/metabolismo , Relación Estructura-Actividad , Receptor Toll-Like 3/metabolismo
8.
EBioMedicine ; 52: 102645, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32014819

RESUMEN

BACKGROUND: TLRs are some of the actively pursued drug-targets in immune disorders. Owing to a recent surge in the cognizance of TLR structural biology and signalling pathways, numerous therapeutic modulators, ranging from low-molecular-weight organic compounds to polypeptides and nucleic acid agents have been developed. METHODS: A penetratin-conjugated small peptide (TIP3), derived from the core ß-sheet of TIRAP, was evaluated in vitro by monitoring the TLR-mediated cytokine induction and quantifying the protein expression using western blot. The therapeutic potential of TIP3 was further evaluated in TLR-dependent in vivo disease models. FINDINGS: TIP3 blocks the TLR4-mediated cytokine production through both the MyD88- and TRIF-dependent pathways. A similar inhibitory-effect was exhibited for TLR3 but not on other TLRs. A profound therapeutic effect was observed in vivo, where TIP3 successfully alleviated the inflammatory response in mice model of collagen-induced arthritis and ameliorated the disease symptoms in psoriasis and SLE models. INTERPRETATION: Our data suggest that TIP3 may be a potential lead candidate for the development of effective therapeutics against TLR-mediated autoimmune disorders. FUNDING: This work was supported by the National Research Foundation of Korea (NRF-2019M3A9A8065098, 2019M3D1A1078940 and 2019R1A6A1A11051471). The funders did not have any role in the design of the present study, data collection, data analysis, interpretation, or the writing of the manuscript.


Asunto(s)
Glicoproteínas de Membrana/química , Péptidos/química , Péptidos/farmacología , Conformación Proteica en Lámina beta , Receptores de Interleucina-1/química , Receptor Toll-Like 4/química , Secuencia de Aminoácidos , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Autoinmunidad , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Modelos Moleculares , Óxido Nítrico/metabolismo , Péptidos/metabolismo , Psoriasis/tratamiento farmacológico , Psoriasis/inmunología , Psoriasis/metabolismo , Psoriasis/patología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like/metabolismo
9.
Semin Cancer Biol ; 64: 61-82, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31054927

RESUMEN

Immune cells of the myeloid and lymphoid lineages express Toll-like receptors (TLRs) to recognize pathogenic components or cellular debris and activate the immune system through the secretion of cytokines. Cytokines are signaling molecules that are structurally and functionally distinct from one another, although their secretion profiles and signaling cascades often overlap. This situation gives rise to pleiotropic cell-to-cell communication pathways essential for protection from infections as well as cancers. Nonetheless, deregulated signaling can have detrimental effects on the host, in the form of inflammatory or autoimmune diseases. Because cytokines are associated with numerous autoimmune and cancerous conditions, therapeutic strategies to modulate these molecules or their biological responses have been immensely beneficial over the years. There are still challenges in the regulation of cytokine function in patients, even in those who take approved biological therapeutics. In this review, our purpose is to discuss the differential expression patterns of TLR-regulated cytokines and their cell type specificity that is associated with cancers and immune-system-related diseases. In addition, we highlight key structural features and molecular recognition of cytokines by receptors; these data have facilitated the development and approval of several biologics for the treatment of autoimmune diseases and cancers.


Asunto(s)
Enfermedades Autoinmunes/terapia , Citocinas/inmunología , Inmunidad Innata/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Receptores Toll-Like/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Citocinas/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal
10.
Exp Mol Med ; 51(4): 1-19, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31028244

RESUMEN

Toll-like receptors (TLRs) recognize pathogen/damage-associated molecular patterns and initiate inflammatory signaling cascades. Occasionally, overexpression of TLRs leads to the onset of numerous inflammatory diseases, necessitating the development of selective inhibitors to allow a protective yet balanced immune response. Here, we demonstrate that a novel peptide (TIP1) derived from Toll/interleukin-1 receptor (TIR) domain-containing adapter protein inhibited multiple TLR signaling pathways (MyD88-dependent and MyD88-independent) in murine and human cell lines. TIP1 also inhibited NLRP3-mediated IL-1ß secretion, as we validated at both the protein and mRNA levels. Biophysical experiments confirmed that TIP1 specifically binds to the BB loop of the TLR4-TIR domain. Animal studies revealed that TIP1 inhibited the secretion of lipopolysaccharide (LPS)-induced proinflammatory cytokines in collagen-induced arthritis (CIA) and kaolin/carrageenan-induced arthritis (K/C) rodent models. TIP1 also rescued animals from sepsis and from LPS-induced kidney/liver damage. Importantly, TIP1 ameliorated the symptoms of rheumatoid arthritis in CIA and K/C rodent models, suggesting that TIP1 has therapeutic potential for the treatment of TLR-mediated autoimmune/inflammatory diseases.


Asunto(s)
Receptores Toll-Like/metabolismo , Animales , Western Blotting , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Interferón beta/metabolismo , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal , Péptidos/farmacología , Células RAW 264.7 , Ratas Sprague-Dawley , Transducción de Señal/genética , Transducción de Señal/fisiología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like/genética
11.
Front Immunol ; 9: 489, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593733

RESUMEN

Toll-like receptors (TLRs) are a unique category of pattern recognition receptors that recognize distinct pathogenic components, often utilizing the same set of downstream adaptors. Specific molecular features of extracellular, transmembrane (TM), and cytoplasmic domains of TLRs are crucial for coordinating the complex, innate immune signaling pathway. Here, we constructed a full-length structural model of TLR4-a widely studied member of the interleukin-1 receptor/TLR superfamily-using homology modeling, protein-protein docking, and molecular dynamics simulations to understand the differential domain organization of TLR4 in a membrane-aqueous environment. Results showed that each functional domain of the membrane-bound TLR4 displayed several structural transitions that are biophysically essential for plasma membrane integration. Specifically, the extracellular and cytoplasmic domains were partially immersed in the upper and lower leaflets of the membrane bilayer. Meanwhile, TM domains tilted considerably to overcome the hydrophobic mismatch with the bilayer core. Our analysis indicates an alternate dimerization or a potential oligomerization interface of TLR4-TM. Moreover, the helical properties of an isolated TM dimer partly agree with that of the full-length receptor. Furthermore, membrane-absorbed or solvent-exposed surfaces of the toll/interleukin-1 receptor domain are consistent with previous X-ray crystallography and biochemical studies. Collectively, we provided a complete structural model of membrane-bound TLR4 that strengthens our current understanding of the complex mechanism of receptor activation and adaptor recruitment in the innate immune signaling pathway.


Asunto(s)
Membrana Dobles de Lípidos/química , Modelos Químicos , Fosfolípidos/química , Receptor Toll-Like 4/química , Biología Computacional , Cristalografía por Rayos X , Dimerización , Humanos , Inmunidad Innata , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Conformación Proteica , Ingeniería de Proteínas , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
12.
Front Immunol ; 9: 75, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434596

RESUMEN

The toll/interleukin 1 receptor (TIR) domain-containing adaptor protein (TIRAP) plays an important role in the toll-like receptor (TLR) 2, TLR4, TLR7, and TLR9 signaling pathways. TIRAP anchors to phosphatidylinositol (PI) 4,5-bisphosphate (PIP2) on the plasma membrane and PI (3,4,5)-trisphosphate (PIP3) on the endosomal membrane and assists in recruitment of the myeloid differentiation primary response 88 protein to activated TLRs. To date, the structure and mechanism of TIRAP's membrane association are only partially understood. Here, we modeled an all-residue TIRAP dimer using homology modeling, threading, and protein-protein docking strategies. Molecular dynamics simulations revealed that PIP2 creates a stable microdomain in a dipalmitoylphosphatidylcholine bilayer, providing TIRAP with its physiologically relevant orientation. Computed binding free energy values suggest that the affinity of PI-binding domain (PBD) for PIP2 is stronger than that of TIRAP as a whole for PIP2 and that the short PI-binding motif (PBM) contributes to the affinity between PBD and PIP2. Four PIP2 molecules can be accommodated by distinct lysine-rich surfaces on the dimeric PBM. Along with the known PI-binding residues (K15, K16, K31, and K32), additional positively charged residues (K34, K35, and R36) showed strong affinity toward PIP2. Lysine-to-alanine mutations at the PI-binding residues abolished TIRAP's affinity for PIP2; however, K34, K35, and R36 consistently interacted with PIP2 headgroups through hydrogen bond (H-bond) and electrostatic interactions. TIRAP exhibited a PIP2-analogous intermolecular contact and binding affinity toward PIP3, aided by an H-bond network involving K34, K35, and R36. The present study extends our understanding of TIRAP's membrane association, which could be helpful in designing peptide decoys to block TLR2-, TLR4-, TLR7-, and TLR9-mediated autoimmune diseases.


Asunto(s)
Membrana Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Fosfatidilinositoles/metabolismo , Receptores de Interleucina-1/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/química , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Membrana Celular/química , Humanos , Glicoproteínas de Membrana/química , Modelos Moleculares , Fosfatidilinositoles/química , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Receptores de Interleucina-1/química
13.
Sci Rep ; 7(1): 11362, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28900197

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) has evolved to navigate through the sophisticated network of a host's immune system. The immune evasion mechanism including type 1 interferon and protein kinase R-mediated antiviral stress responses has been recently attributed to the involvement of MERS-CoV protein 4a (p4a) that masks the viral dsRNA. However, the structural mechanism of how p4a recognizes and establishes contacts with dsRNA is not well explained. In this study, we report a dynamic mechanism deployed by p4a to engage the viral dsRNA and make it unavailable to the host immune system. Multiple variants of p4a-dsRNA were created and investigated through extensive molecular dynamics procedures to highlight crucial interfacial residues that may be used as potential pharmacophores for future drug development. The structural analysis revealed that p4a exhibits a typical αßßßα fold structure, as found in other dsRNA-binding proteins. The α1 helix and the ß1-ß2 loop play a crucial role in recognizing and establishing contacts with the minor grooves of dsRNA. Further, mutational and binding free energy analyses suggested that in addition to K63 and K67, two other residues, K27 and W45, might also be crucial for p4a-dsRNA stability.


Asunto(s)
Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Infecciones por Coronavirus/virología , Enlace de Hidrógeno , Modelos Moleculares , Conformación Molecular , Mutagénesis , Unión Proteica , ARN Bicatenario/genética , ARN Viral/genética , Relación Estructura-Actividad
14.
Molecules ; 21(11)2016 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-27845762

RESUMEN

Toll-like receptors (TLRs) are the most upstream pattern recognition receptors in the cell, which detect pathogen associated molecular patterns and initiate signal transduction, culminating in the transcription of pro-inflammatory cytokines and antiviral interferon. Interleukin-1 receptor-associated kinase 4 (IRAK4) is a key mediator in TLR (except for TLR3) and interleukin-1 receptor signaling pathways. The loss of kinase function of IRAK4 is associated with increased susceptibility to various pathogens, while its over-activation causes autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, and cancer. The therapeutic importance of this master kinase has been advocated by a number of recent preclinical studies, where potent inhibitors have been administered to improve various TLR-mediated pathologies. Increasing studies of X-ray crystallographic structures with bound inhibitors have improved our knowledge on the molecular recognition of ligands by IRAK4, which will be crucial for the development of new inhibitors with improved potencies. In this review, we briefly discuss the structural aspect of ligand recognition by IRAK4 and highlight its therapeutic importance in the context of TLR-associated unmet medical needs.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/química , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Ligandos , Modelos Moleculares , Transducción de Señal
15.
Expert Opin Ther Pat ; 26(6): 719-30, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27136061

RESUMEN

INTRODUCTION: Pattern recognition receptors (PRRs) of the innate immune system mediate and control the activation and progression of adaptive immunity. Toll-like receptors (TLRs) are the most notable of the PRRs: they play crucial roles in protecting the host body against invading pathogens or endogenously released hazardous molecules. Sustained TLR signaling even after the clearance of pathogens or failure to distinguish between 'self' and 'non-self' molecules can cause inflammatory disorders, autoimmune diseases, and cancer. AREAS COVERED: This review focuses on recently developed therapeutic agents with TLR-antagonistic activities. EXPERT OPINION: In recent years, several research institutes and pharmaceutical companies have achieved fundamental successes in inhibiting or reducing TLR signaling and associated effector mechanisms by using novel inhibitors. These inhibitory molecules include antibodies against TLRs, TLR-derived transmembrane (TM) peptides, bacterial-secreted proteins, and natural or synthetic small molecules, peptides, and proteins. Antagonist developers generally target the TLR ectodomain to block receptor activation. The TM and cytosolic Toll/IL-1 receptor domains also have regions that should be explored for the design of peptide-based and small molecule blocking agents. A number of preclinical and clinical breakthroughs may result in the availability of improved TLR immunomodulatory drugs to address important unmet medical needs.


Asunto(s)
Diseño de Fármacos , Factores Inmunológicos/farmacología , Receptores Toll-Like/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/fisiopatología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Patentes como Asunto , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo
16.
Anim Biotechnol ; 27(3): 141-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26953680

RESUMEN

The copy number variation (CNV) is the number of copies of a particular gene in the genotype of an individual. Recent evidences show that the CNVs can vary in frequency and occurrence between breeds. These variations reportedly allowed different breeds to adapt to different environments. As copy number variations follow Mendelian pattern of inheritance, identification and distribution of these variants between populations can be used to infer the evolutionary history of the species. In this study, we have examined the absolute copy number of four Heat shock factor genes viz. HSF-1, 2, 4, and 5 in two different breeds of buffalo species using real-time PCR. Here, we report that the absolute copy number of HSF2 varies between the two breeds. In contrast no significant difference was observed in the copy number for HSF-1, 4, and 5 between the two breeds. Our results provide evidence for the presence of breed specific differences in HSF2 genomic copy number. This seems to be the first step in delineating the genetic factors underlying environmental adaptation between the two breeds. Nevertheless, a more detailed study is needed to characterize the functional consequence of this variation.


Asunto(s)
Búfalos/genética , Variaciones en el Número de Copia de ADN/genética , Proteínas de Choque Térmico/genética , Factores de Transcripción/genética , Animales , ADN/análisis , ADN/genética , Genoma , Masculino , Testículo/química
17.
J Mol Graph Model ; 65: 15-26, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26897084

RESUMEN

Nucleotide binding and oligomerization domain 2 (NOD2), a member of intracellular NOD-like receptors (NLRs) family, recognizes the bacterial peptidoglycan, muramyl dipeptide (MDP) and initiates host immune response. The precise ligand recognition mechanism of NOD2 has remained elusive, although studies have suggested leucine rich repeat (LRR) region of NOD2 as the possible binding site of MDP. In this study, we identified multiple transcripts of NOD2 gene in buffalo (buNOD2) and at least five LRR variants (buNOD2-LRRW (wild type), buNOD2-LRRV1-V4) were found to be expressed in buffalo peripheral blood mononuclear cells. The newly identified buNOD2 transcripts were shorter in lengths as a result of exon-skipping and frame-shift mutations. Among the variants, buNOD2-LRRW, V1, and V3 were expressed more frequently in the animals studied. A comparative receptor-ligand interaction study through modeling of variants, docking, and molecular dynamics simulation revealed that the binding affinity of buNOD2-LRRW towards MDP was greater than that of the shorter variants. The absence of a LRR segment in the buNOD2 variants had probably affected their affinity toward MDP. Notwithstanding a high homology among the variants, the amino acid residues that interact with MDP were located on different LRR motifs. The binding free energy calculation revealed that the amino acids Arg850(LRR4) and Glu932(LRR7) of buNOD2-LRRW, Lys810(LRR3) of buNOD2-LRRV1, and Lys830(LRR3) of buNOD2-LRRV3 largely contributed towards MDP recognition. The knowledge of MDP recognition and binding modes on buNOD2 variants could be useful to understand the regulation of NOD-mediated immune response as well as to develop next generation anti-inflammatory compounds.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/química , Leucocitos Mononucleares/inmunología , Proteína Adaptadora de Señalización NOD2/química , Nucleótidos/química , ARN Mensajero/genética , Acetilmuramil-Alanil-Isoglutamina/inmunología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Búfalos , Exones , Regulación de la Expresión Génica , Intrones , Leucocitos Mononucleares/citología , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/inmunología , Nucleótidos/inmunología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , ARN Mensajero/inmunología , Alineación de Secuencia , Termodinámica
18.
PLoS One ; 10(12): e0144741, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26675301

RESUMEN

Cathelicidins are an ancient class of antimicrobial peptides (AMPs) with broad spectrum bactericidal activities. In this study, we investigated the diversity and biological activity of cathelicidins of buffalo, a species known for its disease resistance. A series of new homologs of cathelicidin4 (CATHL4), which were structurally diverse in their antimicrobial domain, was identified in buffalo. AMPs of newly identified buffalo CATHL4s (buCATHL4s) displayed potent antimicrobial activity against selected Gram positive (G+) and Gram negative (G-) bacteria. These peptides were prompt to disrupt the membrane integrity of bacteria and induced specific changes such as blebing, budding, and pore like structure formation on bacterial membrane. The peptides assumed different secondary structure conformations in aqueous and membrane-mimicking environments. Simulation studies suggested that the amphipathic design of buCATHL4 was crucial for water permeation following membrane disruption. A great diversity, broad-spectrum antimicrobial action, and ability to induce an inflammatory response indicated the pleiotropic role of cathelicidins in innate immunity of buffalo. This study suggests short buffalo cathelicidin peptides with potent bactericidal properties and low cytotoxicity have potential translational applications for the development of novel antibiotics and antimicrobial peptidomimetics.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Catelicidinas/química , Catelicidinas/farmacología , Relación Estructura-Actividad , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/farmacología , Bacterias/efectos de los fármacos , Búfalos , Catelicidinas/clasificación , Catelicidinas/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Dosificación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Mediadores de Inflamación , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Conformación Proteica , Alineación de Secuencia
19.
Mol Biosyst ; 11(8): 2324-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26079944

RESUMEN

Nucleotide-binding and oligomerization domain-containing protein 1 (NOD1) and NOD2 are cytosolic pattern-recognition receptors (PRRs) composed of an N-terminal caspase activation and recruitment domain (CARD), a central NACHT domain and C-terminal leucine-rich repeats (LRRs). They play a vital role in innate immune signaling by activating the NF-κB pathway via recognition of peptidoglycans by LRRs, and ATP-dependent self-oligomerization of NACHT followed by downstream signaling. After oligomerization, CARD/s play a crucial role in activating downstream signaling via the adaptor molecule, RIP2. Due to the inadequacy of experimental 3D structures of CARD/s of NOD2 and RIP2, and results from differential experimental setups, the RIP2-mediated CARD-CARD interaction has remained as a contradictory statement. We employed a combinatorial approach involving protein modeling, docking, molecular dynamics simulation, and binding free energy calculation to illuminate the molecular mechanism that shows the possible involvement of either the acidic or basic patch of zebrafish NOD1/2-CARD/a and RIP2-CARD in CARD-CARD interaction. Herein, we have hypothesized 'type-I' mode of CARD-CARD interaction in NOD1 and NOD2, where NOD1/2-CARD/a involve their acidic surfaces to interact with RIP2. Asp37 and Glu51 (of NOD1) and Arg477, Arg521 and Arg529 (of RIP2) were identified to be crucial for NOD1-RIP2 interaction. However, in NOD2-RIP2, Asp32 (of NOD2) and Arg477 and Arg521 (of RIP2) were anticipated to be significant for downstream signaling. Furthermore, we found that strong electrostatic contacts and salt bridges are crucial for protein-protein interactions. Altogether, our study has provided novel insights into the RIP2-mediated CARD-CARD interaction in zebrafish NOD1 and NOD2, which will be helpful to understand the molecular basis of the NOD1/2 signaling mechanism.


Asunto(s)
Complejos Multiproteicos/química , FN-kappa B/química , Proteína Adaptadora de Señalización NOD1/química , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/química , Proteínas de Pez Cebra/química , Secuencia de Aminoácidos/genética , Animales , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Complejos Multiproteicos/genética , Mutación , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/metabolismo , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
20.
Anim Biotechnol ; 26(3): 211-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25927167

RESUMEN

A novel noninvasive genomic DNA isolation protocol from fecal tissue, by the proteinase K digestion and guanidine hydrochloride extraction method, was assessed for the genotyping of cattle and buffalo. The epithelial tissues present on the surface of the feces were used as source for isolation of genomic DNA. The DNA isolated from fecal tissue was found to be similar as those obtained from other body tissues such as skin, brain, liver, kidney, and muscle. The quality of DNA was checked by agarose gel electrophoresis and polymerase chain reaction (PCR). We successfully amplified a 320 bp MHC class II DRB gene and a 125 bp mt-DNA D-loop region from isolated genomic DNA of cattle. Thus, the DNA isolated using this method was suitable for common molecular biology methods, such as restriction enzyme digestion and genotyping of dairy animals through PCR.


Asunto(s)
ADN/aislamiento & purificación , Células Epiteliales/química , Heces/citología , Animales , Bovinos , ADN/análisis , Femenino , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA