Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.524
Filtrar
Más filtros











Intervalo de año de publicación
1.
PLoS Pathog ; 20(8): e1012463, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39146353

RESUMEN

Circular RNAs (circRNAs) are involved in various physiological and pathological processes in both vertebrates and invertebrates. However, most studies on circRNAs have focused on their roles as endogenous competitive RNAs. Here, we report a novel function of circRNA derived from the Fibrinogen-like protein 1 gene (circ-FGL1) that inhibits coelomocyte apoptosis via competing with the deubiquitinase AjOTUB1 to bind AjMyc in Apostichopus japonicus during Vibrio splendidus infection. The results showed that circ-FGL1 is significantly downregulated in coelomocytes of V. splendidus-induced A. japonicus and negatively regulates coelomocyte apoptosis through the AjBax-AjCyt c pathway. Mechanistically, the deubiquitinase AjOTUB1 and circ-FGL1 could interact with the transcription factor protein AjMyc in the same region with circ-FGL1/AjMyc having greater affinity. Under normal conditions, high levels of circ-FGL1 bind directly to AjMyc, inhibiting the deubiquitylation of AjMyc by AjOTUB1 and leading to the degradation of AjMyc. After V. splendidus infection, AjMyc disassociates from the depressed expression of circ-FGL1, promoting its deubiquitylation by binding to the induced deubiquitinase AjOTUB1 to inhibit its degradation. AjMyc is then transferred to the nucleus and promotes the transcription of AjCyt c and AjBax to induce coelomocyte apoptosis. The new finding will expand our present outstanding on the functional role of circRNAs and suggest new therapeutic targets for the treatment of echinoderms during bacterial invasion.


Asunto(s)
Apoptosis , ARN Circular , Stichopus , Vibriosis , Vibrio , Animales , ARN Circular/metabolismo , ARN Circular/genética , Stichopus/microbiología , Stichopus/metabolismo , Stichopus/genética , Vibriosis/metabolismo , Unión Competitiva , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
2.
Pharmacol Res Perspect ; 12(4): e1223, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39031734

RESUMEN

Fluorescent ligands have proved to be powerful tools in the study of G protein-coupled receptors in living cells. Here we have characterized a new fluorescent ligand PSB603-BY630 that has high selectivity for the human adenosine A2B receptor (A2BR). The A2BR appears to play an important role in regulating immune responses in the tumor microenvironment. Here we have used PSB603-BY630 to monitor specific binding to A2BRs in M1- and M2-like macrophages derived from CD14+ human monocytes. PSB603-BY630 bound with high affinity (18.3 nM) to nanoluciferase-tagged A2BRs stably expressed in HEK293G cells. The ligand exhibited very high selectivity for the A2BR with negligible specific-binding detected at NLuc-A2AR, NLuc-A1R, or NLuc-A3R receptors at concentrations up to 500 nM. Competition binding studies showed the expected pharmacology at A2BR with the A2BR-selective ligands PSB603 and MRS-1706 demonstrating potent inhibition of the specific binding of 50 nM PSB603-BY630 to A2BR. Functional studies in HEK293G cells using Glosensor to monitor Gs-coupled cyclic AMP responses indicated that PSB603-BY630 acted as a negative allosteric regular of the agonist responses to BAY 60-6583. Furthermore, flow cytometry analysis confirmed that PSB603-BY630 could be used to selectively label endogenous A2BRs expressed on human macrophages. This ligand should be an important addition to the library of fluorescent ligands which are selective for the different adenosine receptor subtypes, and will enable study of the role of A2BRs on immune cells in the tumor microenvironment.


Asunto(s)
Colorantes Fluorescentes , Macrófagos , Receptor de Adenosina A2B , Humanos , Células HEK293 , Receptor de Adenosina A2B/metabolismo , Ligandos , Colorantes Fluorescentes/química , Macrófagos/metabolismo , Macrófagos/inmunología , Unión Competitiva , Antagonistas del Receptor de Adenosina A2/farmacología , Agonistas del Receptor de Adenosina A2/farmacología
3.
J Chem Inf Model ; 64(13): 5262-5272, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38869471

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a highly virulent pathogen whose nuclear localization signal (NLS) sequence from capsid protein binds to the host importin-α transport protein and blocks nuclear import. We studied the molecular mechanisms by which two small ligands, termed I1 and I2, interfere with the binding of VEEV's NLS peptide to importin-α protein. To this end, we performed all-atom replica exchange molecular dynamics simulations probing the competitive binding of the VEEV coreNLS peptide and I1 or I2 ligand to the importin-α major NLS binding site. As a reference, we used our previous simulations, which examined noncompetitive binding of the coreNLS peptide or the inhibitors to importin-α. We found that both inhibitors completely abrogate the native binding of the coreNLS peptide, forcing it to adopt a manifold of nonnative loosely bound poses within the importin-α major NLS binding site. Both inhibitors primarily destabilize the native coreNLS binding by masking its amino acids rather than competing with it for binding to importin-α. Because I2, in contrast to I1, binds off-site localizing on the edge of the major NLS binding site, it inhibits fewer coreNLS native binding interactions than I1. Structural analysis is supported by computations of the free energies of the coreNLS peptide binding to importin-α with or without competition from the inhibitors. Specifically, both inhibitors reduce the free energy gain from coreNLS binding, with I1 causing significantly larger loss than I2. To test our simulations, we performed AlphaScreen experiments measuring IC50 values for both inhibitors. Consistent with in silico results, the IC50 value for I1 was found to be lower than that for I2. We hypothesize that the inhibitory action of I1 and I2 ligands might be specific to the NLS from VEEV's capsid protein.


Asunto(s)
Unión Competitiva , Simulación de Dinámica Molecular , Señales de Localización Nuclear , alfa Carioferinas , alfa Carioferinas/metabolismo , alfa Carioferinas/química , alfa Carioferinas/antagonistas & inhibidores , Ligandos , Señales de Localización Nuclear/química , Virus de la Encefalitis Equina Venezolana/metabolismo , Virus de la Encefalitis Equina Venezolana/química , Unión Proteica , Péptidos/química , Péptidos/metabolismo , Péptidos/farmacología , Secuencia de Aminoácidos
4.
J Biol Chem ; 300(6): 107382, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38763337

RESUMEN

ASCT2 (alanine serine cysteine transporter 2), a member of the solute carrier 1 family, mediates Na+-dependent exchange of small neutral amino acids across cell membranes. ASCT2 was shown to be highly expressed in tumor cells, making it a promising target for anticancer therapies. In this study, we explored the binding mechanism of the high-affinity competitive inhibitor L-cis hydroxyproline biphenyl ester (Lc-BPE) with ASCT2, using electrophysiological and rapid kinetic methods. Our investigations reveal that Lc-BPE binding requires one or two Na+ ions initially bound to the apo-transporter with high affinity, with Na1 site occupancy being more critical for inhibitor binding. In contrast to the amino acid substrate bound form, the final, third Na+ ion cannot bind, due to distortion of its binding site (Na2), thus preventing the formation of a translocation-competent complex. Based on the rapid kinetic analysis, the application of Lc-BPE generated outward transient currents, indicating that despite its net neutral nature, the binding of Lc-BPE in ASCT2 is weakly electrogenic, most likely because of asymmetric charge distribution within the amino acid moiety of the inhibitor. The preincubation with Lc-BPE also led to a decrease of the turnover rate of substrate exchange and a delay in the activation of substrate-induced anion current, indicating relatively slow Lc-BPE dissociation kinetics. Overall, our results provide new insight into the mechanism of binding of a prototypical competitive inhibitor to the ASCT transporters.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC , Antígenos de Histocompatibilidad Menor , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/química , Cinética , Antígenos de Histocompatibilidad Menor/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/química , Humanos , Sodio/metabolismo , Sodio/química , Animales , Unión Competitiva
5.
J Thromb Haemost ; 22(8): 2306-2315, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38762021

RESUMEN

BACKGROUND: Vaccine-induced immune thrombotic thrombocytopenia (VITT) is a complication of adenoviral-based vaccine against SARS-CoV-2 due to prothrombotic immunoglobulin (Ig) G antibodies to platelet factor 4 (PF4) and may be difficult to distinguish from heparin-induced thrombocytopenia (HIT) in patients treated with heparin. OBJECTIVES: We assessed the usefulness of competitive anti-PF4 enzyme immunoassays (EIAs) in this context. METHODS: The ability of F(ab')2 fragments of 1E12, 1C12, and 2E1, 3 monoclonal anti-PF4 antibodies, to inhibit the binding of human VITT or HIT antibodies to PF4 was evaluated using EIAs. Alanine-scanning mutagenesis was performed to define the amino acids involved in the interactions between the monoclonal antibodies and PF4. RESULTS: A strong inhibition of VITT IgG binding to PF4 was measured with 1E12 (median inhibition, 93%; n = 8), whereas it had no effect on the binding of HIT antibodies (median, 6%; n = 8). In contrast, 1C12 and 2E1 inhibited VITT (median, 74% and 76%, respectively) and HIT antibodies (median, 68% and 53%, respectively) binding to PF4. When a competitive anti-PF4 EIA was performed with 1E12 for 19 additional VITT samples, it strongly inhibited IgG binding to PF4, except for 1 patient, who had actually developed HIT according to the clinical history. Epitope mapping showed that 1E12 interacts with 5 key amino acids on PF4, of which 4 are also required for the binding of human VITT antibodies, thus explaining the competitive inhibition. CONCLUSION: A simple competitive anti-PF4 EIA with 1E12 could help confirm VITT diagnosis and distinguish it from HIT in patients when both diagnoses are possible.


Asunto(s)
Anticuerpos Monoclonales , Vacunas contra la COVID-19 , Heparina , Factor Plaquetario 4 , Humanos , Factor Plaquetario 4/inmunología , Heparina/efectos adversos , Heparina/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/efectos adversos , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Valor Predictivo de las Pruebas , Anticoagulantes/efectos adversos , Anticoagulantes/inmunología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/diagnóstico , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/efectos adversos , Unión Proteica , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/diagnóstico , Púrpura Trombocitopénica Idiopática/inducido químicamente , SARS-CoV-2/inmunología , Unión Competitiva , Púrpura Trombocitopénica Trombótica/inmunología , Púrpura Trombocitopénica Trombótica/diagnóstico , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/inducido químicamente
6.
Toxicol Mech Methods ; 34(5): 563-571, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38317456

RESUMEN

BACKGROUND: In a previous study, diethylstilbestrol (DES) was shown to induce oocyte maturation in fish. In the present study, the interaction of DES on goldfish membrane progesterone receptor α (GmPRα) was investigated using a competitive binding assay with radiolabeled steroids. The results indicate that DES exerts its effects on membrane progesterone receptor alpha (mPRα) and induces oocyte maturation through nongenomic steroid mechanisms. This study provides empirical data that demonstrate the binding between DES and GmPRα. METHODS: Binding of DES to GmPRα was achieved by using radiolabeled DES and recombinant GmPRα expressed in culture cells or purified GmPRα proteins that coupled to graphene quantum dots (GQDs). Additionally, the competitive binding of fluorescently labeled progesterone to GmPRα-expressing cells was evaluated. RESULTS: Although significant nonspecific binding of radiolabeled DES to the cell membrane that expresses GmPRα has been observed, specific binding of DES to GmPRα has been successfully identified in the presence of digitonin. Furthermore, the specific binding of DES to GmPRα was confirmed by a binding assay using GQD-GmPRα. The radiolabeled DES was shown to bind to GQD-GmPRα. Additionally, the competition for the binding of fluorescently labeled progesterone to GmPRα-expressing cells was achieved with the DES. CONCLUSIONS: The results of the experiments revealed that DES binds to GmPRα. Thus, it can be concluded that DES induces goldfish oocyte maturation by binding to GmPRα.


Asunto(s)
Dietilestilbestrol , Proteínas de Peces , Carpa Dorada , Receptores de Progesterona , Animales , Unión Competitiva , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Dietilestilbestrol/toxicidad , Proteínas de Peces/metabolismo , Proteínas de Peces/genética , Carpa Dorada/metabolismo , Oocitos/metabolismo , Oocitos/efectos de los fármacos , Progesterona/metabolismo , Unión Proteica , Receptores de Progesterona/metabolismo
7.
Mol Pharmacol ; 105(4): 301-312, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38346795

RESUMEN

Atypical chemokine receptor 3 (ACKR3), formerly referred to as CXCR7, is considered to be an interesting drug target. In this study, we report on the synthesis, pharmacological characterization and radiolabeling of VUF15485, a new ACKR3 small-molecule agonist, that will serve as an important new tool to study this ß-arrestin-biased chemokine receptor. VUF15485 binds with nanomolar affinity (pIC50 = 8.3) to human ACKR3, as measured in [125I]CXCL12 competition binding experiments. Moreover, in a bioluminescence resonance energy transfer-based ß-arrestin2 recruitment assay VUF15485 acts as a potent ACKR3 agonist (pEC50 = 7.6) and shows a similar extent of receptor activation compared with CXCL12 when using a newly developed, fluorescence resonance energy transfer-based ACKR3 conformational sensor. Moreover, the ACKR3 agonist VUF15485, tested against a (atypical) chemokine receptor panel (agonist and antagonist mode), proves to be selective for ACKR3. VUF15485 labeled with tritium at one of its methoxy groups ([3H]VUF15485), binds ACKR3 saturably and with high affinity (K d = 8.2 nM). Additionally, [3H]VUF15485 shows rapid binding kinetics and consequently a short residence time (<2 minutes) for binding to ACKR3. The selectivity of [3H]VUF15485 for ACKR3, was confirmed by binding studies, whereupon CXCR3, CXCR4, and ACKR3 small-molecule ligands were competed for binding against the radiolabeled agonist. Interestingly, the chemokine ligands CXCL11 and CXCL12 are not able to displace the binding of [3H]VUF15485 to ACKR3. The radiolabeled VUF15485 was subsequently used to evaluate its binding pocket. Site-directed mutagenesis and docking studies using a recently solved cryo-EM structure propose that VUF15485 binds in the major and the minor binding pocket of ACKR3. SIGNIFICANCE STATEMENT: The atypical chemokine receptor atypical chemokine receptor 3 (ACKR3) is considered an interesting drug target in relation to cancer and multiple sclerosis. The study reports on new chemical biology tools for ACKR3, i.e., a new agonist that can also be radiolabeled and a new ACKR3 conformational sensor, that both can be used to directly study the interaction of ACKR3 ligands with the G protein-coupled receptor.


Asunto(s)
Quimiocina CXCL12 , Receptores CXCR4 , Humanos , Receptores CXCR4/metabolismo , Quimiocina CXCL12/metabolismo , Quimiocina CXCL11/metabolismo , Transducción de Señal , Ligandos , Unión Competitiva
8.
J Am Chem Soc ; 146(1): 187-200, 2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38118119

RESUMEN

The affinity and selectivity of small molecules for proteins drive drug discovery and development. We report a fluorescent probe cellular binding assay (FPCBA) for determination of these values for native (untagged) proteins overexpressed in living cells. This method uses fluorophores such as Pacific Blue (PB) linked to cell-permeable protein ligands to generate probes that rapidly and reversibly equilibrate with intracellular targets, as established by kinetic assays of cellular uptake and efflux. To analyze binding to untagged proteins, an internal ribosomal entry site (IRES) vector was employed that allows a single mRNA to encode both the protein target and a separate orthogonal fluorescent protein (mVenus). This enabled cellular uptake of the probe to be correlated with protein expression by flow cytometry, allowing measurement of cellular dissociation constants (Kd) of the probe. This approach was validated by studies of the binding of allosteric activators to eight different Protein Kinase C (PKC) isozymes. Full-length PKCs expressed in transiently transfected HEK293T cells were used to measure cellular Kd values of a probe comprising PB linked to the natural product phorbol via a carbamate. These values were further used to determine competitive binding constants (cellular Ki values) of the nonfluorescent phorbol ester PDBu and the anticancer agent bryostatin 1 for each isozyme. For some PKC-small molecule pairs, these cellular Ki values matched known biochemical Ki values, but for others, altered selectivity was observed in cells. This approach can facilitate quantification of interactions of small molecules with physiologically relevant native proteins.


Asunto(s)
Ésteres del Forbol , Proteína Quinasa C , Humanos , Células HEK293 , Proteína Quinasa C/química , Unión Competitiva
9.
Acta Biochim Pol ; 70(4): 807-815, 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37934513

RESUMEN

OBJECTIVE: Circular RNAs (circRNAs) are enriched in the brain and involved in various central nervous system diseases. The potential role of circCCDC6 in cerebral ischemia-reperfusion defects was partly elucidated in the work. METHODS: A middle cerebral artery occlusion/reperfusion (MCAO/R) rat model and an oxygen-glucose deprivation and re-oxygenation (OGD/R)-treated SH-SY5Y cell model were constructed. CircCCDC6 expression in the two models was examined, and circCCDC6-involved mechanisms in neuronal pyroptosis and inflammation were analyzed through loss- and gain-of-function assays. RESULTS: MCAO/R rat brain tissues and OGD/R-treated SH-SY5Y cells exhibited upregulated circCCDC6. Silencing circCCDC6 attenuated neuronal pyroptosis and inflammation in the brain tissue of MCAO/R rats. Overexpressing circCCDC6 or inhibiting miR-128-3p stimulated OGD/R-induced pyroptosis and inflammation in SH-SY5Y cells, while upregulating miR-128-3p attenuated OGD/R injury. CircCCDC6 silencing-induced effects on SH-SY5Y cells were antagonized by TXNIP overexpression. CONCLUSION: Mechanistically, circCCDC6 mediates miR-128-3p and activates TXNIP/NLRP3, thereby promoting OGD/R-induced neuronal pyroptosis and inflammation. CircCCDC6 may provide a new strategy for the treatment of MCAO/R.


Asunto(s)
Isquemia Encefálica , MicroARNs , Neuroblastoma , Daño por Reperfusión , Animales , Humanos , Ratas , Apoptosis , Unión Competitiva , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Glucosa/farmacología , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Inflamación , MicroARNs/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Reperfusión , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
10.
Arch Biochem Biophys ; 747: 109753, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37714251

RESUMEN

The MF30 monoclonal antibody, which binds to the myosin subfragment-2 (S2), was found to increase the extent of myofibril shortening. Yet, previous observations found no effect of this antibody on actin sliding over myosin during in vitro motility assays with purified proteins in which myosin binding protein C (MyBPC) was absent. MF30 is hypothesized to enhance the availability of myosin heads (subfragment-1 or S1) to bind actin by destabilizing the myosin S2 coiled-coil and sterically blocking S2 from binding S1. The mechanism of action likely includes MF30's substantial size, thereby inhibiting S1 heads and MyBPC from binding S2. Hypothetically, MF30 should enhance the ON state of myosin, thereby increasing muscle contraction. Our findings indicate that MF30 binds preferentially to the unfolded heavy chains of S2, displaying positive cooperativity. However, the dose-response curve of MF30's enhancement of myofibril shortening did not suggest complex interactions with S2. Single, double, and triple-stained myofibrils with increasing amounts of antibodies against myosin rods indicate a possible competition with MyBPC. Additional assays revealed decreased fluorescence intensity at the C-zone (central zone in the sarcomere, where MyBPC is located), where MyBPC may inhibit MF30 binding. Another monoclonal antibody named MF20, which binds to the light meromyosin (LMM) without affecting myofibril contraction, showed less reduction in fluorescence intensity at the C-zone in expansion microscopy than MF30. Expansion microscopy images of myofibrils labeled with MF20 revealed labeling of the A-band (anisotropic band) and a slight reduction in the labeling at the C-zone. The staining pattern obtained from the expansion microscopy image was consistent with images from photolocalization microscopy which required the synthesis of unique photoactivatable quantum dots, and Zeiss Airyscan imaging as well as alternative expansion microscopy digestion methods. Consistent with the hypothesis that MF30 competes with MyBPC binding to S2, cardiac tissue from MyBPC knockout mice was stained more intensely, especially in the C-zone, by MF30 compared to the wild type.


Asunto(s)
Actinas , Microscopía , Animales , Ratones , Actinas/metabolismo , Unión Competitiva , Miosinas/metabolismo , Subfragmentos de Miosina/metabolismo , Anticuerpos Monoclonales
11.
Int J Oncol ; 63(1)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37264959

RESUMEN

Long noncoding RNAs (lncRNAs) are a type of regulatory molecule with potential roles in the development of several different malignancies. However, the underlying mechanisms of lncRNAs in colorectal cancer (CRC) are incompletely understood. The present study investigated the molecular mechanism of LINC02038 in CRC. LINC02038 expression was decreased in CRC tissues compared to the para­cancerous tissues and LINC02038 overexpression markedly reduced the proliferation, vitality, migration and invasive ability and greatly accelerated apoptosis of colorectal cancer cells. Bioinformatics examination indicated that LINC02038 may have targeted microRNA (miR)­552­5p. RNA immunoprecipitation and luciferase reporter assays showed that LINC02038 served as a sponge for miR­552­5p, hindering target gene FAM172A of miR­552­5p degradation. Moreover, methylated RNA immunoprecipitation (MeRIP)­qualitative PCR assays revealed that YTHDF2 could identify and regulate the METTL3­mediated LINC02038 N6­methyladenosine (m6A) modification and increase its degradation, thereby promoting CRC progression via the PI3K/AKT pathway. Based on the CRC clinical specimens, it was shown that LINC02038 was negatively associated with lymphatic metastasis and distant metastasis. These results revealed that m6A/LINC02038/miR­552­5p/FAM172A may be a novel anti­tumor axis and LINC02038 may serve as a biomarker and treatment option for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , ARN Largo no Codificante , Humanos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Celular Tumoral , Unión Competitiva , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Colorrectales/patología , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Metiltransferasas/metabolismo , Proteínas/genética
12.
Am J Transplant ; 23(7): 920-934, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37054890

RESUMEN

End-stage organ failure often requires solid organ transplantation. Nevertheless, transplant rejection remains an unresolved issue. The induction of donor-specific tolerance is the ultimate goal in transplantation research. In this study, an allograft vascularized skin rejection model using BALB/c-C57/BL6 mice was established to evaluate the regulation of the poliovirus receptor signaling pathway using CD226 knockout or T cell immunoglobulin and ITIM domain (TIGIT)-crystallizable fragment (Fc) recombinant protein treatment. In the TIGIT-Fc-treated and CD226 knockout groups, graft survival time prolonged significantly, with a regulatory T cell proportion increase and M2-type macrophage polarization. Donor-reactive recipient T cells became hyporesponsive while responding normally after a third-party antigen challenge. In both groups, serum interleukin (IL)-1ß, IL-6, IL-12p70, IL-17A, tumor necrosis factor-α, interferon gamma, and monocyte chemoattractant protein-1 levels decreased, and the IL-10 level increased. In vitro, M2 markers, such as Arg1 and IL-10, were markedly increased by TIGIT-Fc, whereas iNOS, IL-1ß, IL-6, IL-12p70, tumor necrosis factor-α, and interferon gamma levels decreased. CD226-Fc exerted the opposite effect. TIGIT suppressed TH1 and TH17 differentiation by inhibiting macrophage SHP-1 phosphorylation and enhanced ERK1/2-MSK1 phosphorylation and nuclear translocation of CREB. In conclusion, CD226 and TIGIT competitively bind to poliovirus receptor with activating and inhibitory functions, respectively. Mechanistically, TIGIT promotes IL-10 transcription from macrophages by activating the ERK1/2-MSK1-CREB pathway and enhancing M2-type polarization. CD226/TIGIT-poliovirus receptor are crucial regulatory molecules of allograft rejection.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T , Rechazo de Injerto , Macrófagos , Receptores Inmunológicos , Trasplante de Piel , Animales , Ratones , Antígenos de Diferenciación de Linfocitos T/metabolismo , Unión Competitiva , Rechazo de Injerto/etiología , Interferón gamma , Interleucina-10 , Interleucina-6 , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa
13.
Nucleic Acids Res ; 51(9): 4363-4384, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36942481

RESUMEN

Crossing over between homologs is critical for the stable segregation of chromosomes during the first meiotic division. Saccharomyces cerevisiae Mer3 (HFM1 in mammals) is a SF2 helicase and member of the ZMM group of proteins, that facilitates the formation of the majority of crossovers during meiosis. Here, we describe the structural organisation of Mer3 and using AlphaFold modelling and XL-MS we further characterise the previously described interaction with Mlh1-Mlh2. We find that Mer3 also forms a previously undescribed complex with the recombination regulating factors Top3 and Rmi1 and that this interaction is competitive with Sgs1BLM helicase. Using in vitro reconstituted D-loop assays we show that Mer3 inhibits the anti-recombination activity of Sgs1 helicase, but only in the presence of Dmc1. Thus we provide a mechanism whereby Mer3 interacts with a network of proteins to protect Dmc1 derived D-loops from dissolution.


Asunto(s)
ADN Helicasas , Recombinación Homóloga , Meiosis , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Ciclo Celular/genética , Intercambio Genético , ADN Helicasas/química , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Meiosis/genética , Unión Proteica , Pliegue de Proteína , RecQ Helicasas/antagonistas & inhibidores , RecQ Helicasas/química , RecQ Helicasas/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Unión Competitiva
14.
Food Res Int ; 164: 112368, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36737955

RESUMEN

Whey protein hydrolysate (WPH), maltodextrin (MD), low methoxy pectin (LMP) and high methoxy pectin (HMP) were used to study the interface binding under high temperature sterilization conditions (121 °C, 15 min). The effect of competitive binding of MD and pectin with interface protein on the storage stability and gastrointestinal fate of fish oil emulsion was studied. The low-molecular-weight MD and the interface protein undergo a wide range of covalent binding through the Maillard reaction, while a small amount of high-molecular-weight pectin can form a protective shell with the interface protein through electrostatic interaction to inhibit the covalent reaction of MD, which was called competitive binding. However, due to the bridging and depletion flocculation of pectin, the emulsification stability of fish oil emulsion reduced. After 13 days of storage, compared with the particle size of the WPH fish oil emulsion (459.18 nm), the fish oil emulsion added with LMP and HMP reached 693.58 nm and 838.54 nm, respectively. In vitro digestion proved that WPH fish oil emulsion flocculated rapidly in the stomach (1.76 µm), while WPH-MD and WPH-MD-pectin fish oil emulsions flocculated slightly (less than800 nm). WPH-MD-pectin delayed digestion in the gastrointestinal tract, and HMP exhibited a better slow-release effect. This study provides reference for the design of multi-component functional drinks and other bioactive ingredient delivery system.


Asunto(s)
Aceites de Pescado , Pectinas , Emulsiones/metabolismo , Proteína de Suero de Leche , Temperatura , Unión Competitiva , Digestión
15.
Int J Biol Macromol ; 231: 123224, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36649871

RESUMEN

Triiodothyronine (T3) and thyroxine (T4) are essential for regulating cell metabolic rate and promoting the development and differentiation of brain tissue, especially in fetuses and newborns. In particular, it has been proved that MeO-PCBs have high binding to thyroid hormone transporters and can competitively bind to thyroid carrier proteins, thus destroying the transport of the thyroid hormone. Fluorescence competition binding experiments and docking results showed that the binding affinity decreased with the increase in number of chlorine atoms of MeO-PCBs. The interaction mechanism of MeO-PCBs with thyroid transporter (TTR) and thyroid binding globulin (TBG) was compared by computational simulation and the binding free energies were calculated by the molecular mechanics/Poisson-Boltzmann surface area (MM/PBSA) method. Electrostatic potential analysis, Hirshfeld surface analysis and electron density difference maps confirmed the existence of electrostatic interactions. Secondly, noncovalent interaction (NCI) analysis further indicated that the main driving force for the combination of MeO-PCBs to TTR and TBG were electrostatic interaction and van der Waals interaction. The conformational changes of the protein after binding were studied by a molecular dynamic simulation.


Asunto(s)
Bifenilos Policlorados , Recién Nacido , Humanos , Bifenilos Policlorados/metabolismo , Unión Competitiva , Prealbúmina , Hormonas Tiroideas/metabolismo , Tiroxina/metabolismo , Proteínas de Transporte de Membrana/metabolismo
16.
J Fluoresc ; 33(1): 13-24, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36209269

RESUMEN

Flavonoids have potential applications in the nutraceutical, medicinal, pharmaceutical and cosmetic fields. The binding of flavonoids with DNA could unravel essential information required for the design of novel and effective chemical agents. The present paper describes the interaction of a flavonoid and a potent anticancer drug, galangin (GAL) with calf thymus DNA (ct-DNA) by fluorescence, UV absorption, melting studies, viscosity measurements and molecular docking studies. A hyperchromic effect was noticed in the absorption spectra of ct-DNA in the presence of the GAL system, indicating the presence of a groove mode of binding. Furthermore, GAL persuaded the minor changes in ct-DNA viscosity, indicating a non-intercalative mode of binding. Fluorescence studies revealed that the GAL quenched the fluorescence intensity of ct-DNA-Hoechst, thereby indicating the interaction between GAL and ct-DNA. Fluorescence results obtained at 298, 308 and 318 K revealed that the fluorescence quenching of ct-DNA-Hoechst-GAL occurred through the static quenching mechanism. Thermodynamic parameters for ct-DNA-Hoechst-GAL were computed and suitable conclusions were drawn. The changes noticed in the conformation of ct-DNA upon interaction with GAL were evaluated in terms of molar ellipticity. It indicated a plausible interaction between ct-DNA and GAL. The molecular docking studies also confirmed the groove mode of binding in the ct-DNA-GAL system. Thus, this work helped to unravel the binding mechanism between GAL and ct-DNA.


Asunto(s)
ADN , Flavonoides , Simulación del Acoplamiento Molecular , Espectrometría de Fluorescencia , Unión Competitiva , Conformación de Ácido Nucleico , ADN/química , Termodinámica , Dicroismo Circular , Viscosidad , Espectrofotometría Ultravioleta
17.
Mol Cancer Res ; 21(2): 127-139, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36346305

RESUMEN

Anoikis resistance is a prerequisite for circulating tumor cells to survive. However, the mechanism underlying anoikis resistance is poorly understood. In the current study, the effect of TNF receptor-associated factor 6 (TRAF6)-induced NF-kB activation on anoikis susceptibility in tumor cells was evaluated. Differential TRAF6-binding proteins in anoikis-sensitive versus anoikis-resistant tumor cells were screened by LC/MS-MS analysis. The effects of TRAF6-binding proteins on the stability of TRAF6, the activation of NF-kB signaling and anoikis susceptibility in tumor cells were detected. We found that the loss of TRAF6 expression is an important molecular event linked to anoikis. X-linked inhibitor of apoptosis protein (XIAP), an E3 ligase, can bind, ubiquitinate, and degrade TRAF6 and may lead to inactivation of NF-κB signaling and anoikis sensitivity. High expression of prohibitin 1 (PHB1) competes with XIAP for binding to TRAF6 and confers anoikis resistance to tumor cells. PHB1 and TRAF6 knockdown eliminated tumor cells from the circulation in vivo. Significant correlations between elevated PHB1 and TRAF6 expression and distant metastasis were observed in patients with oral cancer. Collectively, we elucidated a novel mechanism governing anoikis. Our data also indicated that TRAF6 and PHB1 are potential therapeutic targets for tumor cells disseminating in the circulation. IMPLICATIONS: Our data implicate that PHB1 competes with XIAP for binding to TRAF6 and confers anoikis resistance to tumor cells.


Asunto(s)
FN-kappa B , Factor 6 Asociado a Receptor de TNF , Humanos , FN-kappa B/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Anoicis/fisiología , Unión Competitiva , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
18.
J Inorg Biochem ; 238: 112043, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36370502

RESUMEN

Protein crystallography and biochemical assays reveal that the organometallic drug, [Ru(η6-p-cymene)Cl2(pta)] (RAPTA-C), preferentially binds to nucleosomal histone proteins in chromatin. To better understand the binding mechanism we report here a mass spectrometric-based competitive binding study between a model peptide from the acidic patch region of the H2A histone protein (the region where RAPTA-C is known to bind) and an oligonucleotide. In contrast to the protein crystallography and biochemical assays, RAPTA-C preferentially binds to the oligonucleotide, confirming that steric factors, rather than electronic effects, primarily dictate binding of RAPTA-C to histone proteins within the nucleosome.


Asunto(s)
Histonas , Compuestos Organometálicos , Histonas/metabolismo , Oligonucleótidos , Unión Competitiva , Compuestos Organometálicos/química , Péptidos/metabolismo
19.
Cell Cycle ; 22(2): 213-228, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35980157

RESUMEN

The present study aimed to assess the role of the long non-coding RNA-urothelial cancer associated 1 (lncRNA-UCA1)/microRNA (miR)-383/vascular endothelial growth factor A (VEGFA) axis in regulating lung adenocarcinoma physiology through in vivo and in vitro experiments. The expression profile of lncRNA-UCA1 was analyzed by genome-wide analysis from GSE146459. The cell counting Kit-8, colony formation, wound healing and transwell assays were performed to evaluate the effects of lncRNA-UCA1 in vitro. In addition, luciferase reporter assays were performed to confirm the binding site. The expression levels of miR-383 and VEGFA in tumor cells were measured using reverse transcription-quantitative PCR. HCC-78 was also transfected with miR-383 mimics, inhibitors and siRNA-VEGFA before their viability was also assessed. Xenograft models were established in nude mice to investigate the tumor characteristics in vivo. The expression of lncRNA-UCA1 was significantly increased in tumor tissues and cells compared with adjacent tissues or HBE cells. Silencing lncRNA-UCA1 expression in cells resulted in a reduction in lung cancer cell viability. In addition, lncRNA-UCA1 silencing increased the expression of miR-383. Inhibiting miR-383 expression increased HCC-78 proliferation, migration and invasion, whilst reducing their apoptosis. miR-383 was shown to specifically target VEGFA to inhibit its expression at both the protein and mRNA levels. VEGFA knockdown resulted in a reduction in all aforementioned aspects of HCC-78 cell activity. In addition, inhibiting miR-383 expression led to larger tumor sizes in vivo. To conclude, the results of the study suggest that lncRNA-UCA1 can regulate the expression of miR-383 and, in turn, VEGFA.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , ARN Largo no Codificante , Neoplasias de la Vejiga Urinaria , Animales , Ratones , Humanos , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Carcinoma Hepatocelular/genética , Unión Competitiva , Ratones Desnudos , Neoplasias Hepáticas/genética , Neoplasias de la Vejiga Urinaria/patología , Pulmón/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
20.
MAbs ; 14(1): 2133674, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36224723

RESUMEN

Therapeutic monoclonal antibodies (mAbs) have a propensity to host a large number of chemical and enzymatical modifications that need to be properly assessed for their potential impact on target binding. Traditional strategies of assessing the criticality of these attributes often involve a laborious and low-throughput variant enrichment step prior to binding affinity measurement. Here, we developed a novel competitive binding-based enrichment strategy followed by mass spectrometry analysis (namely, competitive binding-MS) to achieve high-throughput evaluation of potential critical quality attributes in therapeutic mAbs. Leveraging the differences in target binding capability under competitive binding conditions, the criticality of multiple mAb attributes can be simultaneously evaluated by quantitative mass spectrometry analysis. The utility of this new workflow was demonstrated in three mAb case studies, where different post-translational modifications occurring within the complementarity-determining regions were successfully interrogated for their impact on antigen binding. As this workflow does not require prior enrichment (e.g., by forced degradation or liquid chromatography fractionation) of the variants, it is particularly valuable during the mAb candidate developability assessment, where fast turn-around time is highly desired to assist candidate selection.Abbreviations: ACN: acetonitrile; ADCC: antibody-dependent cell-mediated cytotoxicity; AEX: anion exchange chromatography; bsAb: bispecific antibody; CDC: complement-dependent cytotoxicity; CDR: complementarity-determining region; CML: carboxymethylation; CQA: critical quality attribute; DDA: data-dependent acquisition; DMSO: dimethyl sulfoxide; DTT: dithiothreitol; FA: formic acid; Fab: Fragment antigen-binding; FcRn: neonatal Fc receptor; HC: heavy chain; HIC: hydrophobic interaction chromatography; IAA: iodoacetamide; IEX: ion exchange chromatography; LC: light chain; mAb monoclonal antibody; msAb: monospecific antibody; MS: mass spectrometry; PBS: phosphate-buffered saline; pI: isoelectric point; PTM: post-translational modification; SCX: strong cation exchange chromatography; SEC: size exclusion chromatography; SPR: surface plasmon resonance; XIC: extracted ion chromatography.


Asunto(s)
Anticuerpos Monoclonales , Antineoplásicos Inmunológicos , Acetonitrilos , Anticuerpos Monoclonales/química , Unión Competitiva , Cationes , Regiones Determinantes de Complementariedad/química , Dimetilsulfóxido , Ditiotreitol , Yodoacetamida , Espectrometría de Masas/métodos , Fosfatos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA