Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Life Sci Alliance ; 7(8)2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38782603

RESUMO

It is known that stress influences immune cell function. The underlying molecular mechanisms are unclear. We recently reported that many chemokine receptors (CRs) heteromerize with α1-adrenoceptors (α1-ARs) through which CRs are regulated. Here, we show that arginine vasopressin receptor 1A (AVPR1A) heteromerizes with all human CRs, except chemokine (C-X-C motif) receptor (CXCR)1, in recombinant systems and that such heteromers are detectable in THP-1 cells and human monocytes. We demonstrate that ligand-free AVPR1A differentially regulates the efficacy of CR partners to mediate chemotaxis and that AVPR1A ligands disrupt AVPR1A:CR heteromers, which enhances chemokine (C-C motif) receptor (CCR)1-mediated chemotaxis and inhibits CCR2-, CCR8-, and CXCR4-mediated chemotaxis. Using bioluminescence resonance energy transfer to monitor G protein activation and CRISPR/Cas9 gene-edited THP-1 cells lacking AVPR1A or α1B-AR, we show that CRs that share the propensity to heteromerize with α1B/D-ARs and AVPR1A exist and function within interdependent hetero-oligomeric complexes through which the efficacy of CRs to mediate chemotaxis is controlled. Our findings suggest that hetero-oligomers composed of CRs, α1B/D-ARs, and AVPR1A may enable stress hormones to regulate immune cell trafficking.


Assuntos
Quimiotaxia , Monócitos , Receptores de Quimiocinas , Receptores de Vasopressinas , Humanos , Monócitos/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores de Quimiocinas/genética , Receptores de Vasopressinas/metabolismo , Receptores de Vasopressinas/genética , Células THP-1 , Multimerização Proteica , Células HEK293 , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Sistemas CRISPR-Cas , Transdução de Sinais , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 1/genética , Ligantes
2.
Inorg Chem ; 63(7): 3506-3515, 2024 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-38311840

RESUMO

ZSM-5 zeolites with modified acidity and diffusivity are employed as catalysts for the shape-selective alkylation of toluene with ethanol to para-ethyltoluene (p-ET). To avoid pore blocking and loss of active sites caused by traditional methods of enhancing para-selectivity using modifiers, here, we constructed twin intergrowth structured ZSM-5 (Z5-T), achieving modulation of the inherent acidity and diffusivity through interface engineering. The characterization results demonstrate that due to the intergrowth interface, the Z5-T catalyst forms more inherent Lewis acid sites and also renders more sinusoidal channels opened to the surface. Z5-T with an appropriate acidity and enhanced shape-selectivity inhibits side reactions such as isomerization and coke formation, demonstrating improved p-ET selectivity (>90%) and catalytic stability (>200 h) in the alkylation of toluene with ethanol.

3.
Cells ; 12(18)2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37759456

RESUMO

Long-term alcohol consumption leads to cardiac arrhythmias including atrial fibrillation (AF), the most common alcohol-related arrhythmia. While AF significantly increases morbidity and mortality in patients, it takes years for an alcoholic individual undergoing an adaptive status with normal cardiac function to reach alcoholic cardiomyopathy. The underlying mechanism remains unclear to date. In this study, we assessed the functional role of JNK2 in long-term alcohol-evoked atrial arrhythmogenicity but preserved cardiac function. Wild-type (WT) mice and cardiac-specific JNK2dn mice (with an overexpression of inactive dominant negative (dn) JNK2) were treated with alcohol (2 g/kg daily for 2 months; 2 Mo). Confocal Ca2+ imaging in the intact mouse hearts showed that long-term alcohol prolonged intracellular Ca2+ transient decay, and increased pacing-induced Ca2+ waves, compared to that of sham controls, while cardiac-specific JNK2 inhibition in JNK2dn mice precluded alcohol-evoked Ca2+-triggered activities. Moreover, activated JNK2 enhances diastolic SR Ca2+ leak in 24 h and 48 h alcohol-exposed HL-1 atrial myocytes as well as HEK-RyR2 cells (inducible expression of human RyR2) with the overexpression of tGFP-tagged active JNK2-tGFP or inactive JNK2dn-tGFP. Meanwhile, the SR Ca2+ load and systolic Ca2+ transient amplitude were both increased in ventricular myocytes, along with the preserved cardiac function in 2 Mo alcohol-exposed mice. Moreover, the role of activated JNK2 in SR Ca2+ overload and enhanced transient amplitude was also confirmed in long-term alcohol-exposed HL-1 atrial myocytes. In conclusion, our findings suggest that long-term alcohol-activated JNK2 is a key driver in preserved cardiac function, but at the expense of enhanced cardiac arrhythmogenicity. Modulating JNK2 activity could be a novel anti-arrhythmia therapeutic strategy.


Assuntos
Fibrilação Atrial , Humanos , Animais , Camundongos , Canal de Liberação de Cálcio do Receptor de Rianodina , Etanol/efeitos adversos , Miócitos Cardíacos , Proteínas Quinases JNK Ativadas por Mitógeno , Isoformas de Proteínas
4.
FEBS Lett ; 597(15): 2017-2027, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37395117

RESUMO

It is unknown whether heteromerization between chemokine (C-X-C motif) receptor 4 (CXCR4), atypical chemokine receptor 3 (ACKR3) and α1b -adrenoceptor (α1b -AR) influences effects of the CXCR4/ACKR3 agonist chemokine (C-X-C motif) ligand 12 (CXCL12) and the noncognate CXCR4 agonist ubiquitin on agonist-promoted G protein activation. We provide biophysical evidence that both ligands stimulate CXCR4-mediated Gαi activation. Unlike CXCL12, ubiquitin fails to recruit ß-arrestin. Both ligands differentially modulate the conformation of CXCR4:ACKR3 heterodimers and its propensity to hetero-trimerize with α1b -AR. CXCR4:ACKR3 heterodimerization reduces the potency of CXCL12, but not of ubiquitin, to activate Gαi. Ubiquitin enhances phenylephrine-stimulated α1b -AR-promoted Gαq activation from hetero-oligomers comprising CXCR4. CXCL12 enhances phenylephrine-stimulated α1b -AR-promoted Gαq activation from CXCR4:α1b -AR heterodimers and reduces phenylephrine-stimulated α1b -AR-promoted Gαq activation from ACKR3 comprising heterodimers and trimers. Our findings suggest heteromer and ligand-dependent functions of the receptor partners.


Assuntos
Receptores CXCR4 , Receptores CXCR , Ligantes , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Quimiocina CXCL12/metabolismo , Fenilefrina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Ubiquitina/metabolismo , Receptores Adrenérgicos/metabolismo
5.
PLoS One ; 18(4): e0284472, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37071651

RESUMO

Systemic concentrations of chemokine CCL2, an agonist at chemokine receptors CCR2/3/5, have been associated with hemodynamic instability after traumatic-hemorrhagic shock. We reported previously that the CCR2 antagonist INCB3284 prevents cardiovascular collapse and reduces fluid requirements after 30min of hemorrhagic shock (HS), whereas the CCR5 antagonist Maraviroc was ineffective. The effects of CCR3 blockade after HS are unknown and information on the therapeutic potential of INCB3284 after longer periods of HS and in HS models in the absence of fluid resuscitation (FR) is lacking. The aims of the present study were to assess the effects of CCR3 blockade with SB328437 and to further define the therapeutic efficacy of INCB3284. In series 1-3, Sprague-Dawley rats were hemorrhaged to a mean arterial blood pressure (MAP) of 30mmHg, followed by FR to MAP of 60mmHg or systolic blood pressure of 90mmHg. Series 1: 30min HS and FR until t = 90min. SB328437 at t = 30min dose-dependently reduced fluid requirements by >60%. Series 2: 60min HS and FR until t = 300min. INCB3284 and SB328437 at t = 60min reduced fluid requirements by more than 65% (p<0.05 vs. vehicle) and 25% (p>0.05 vs. vehicle), respectively, until t = 220min. Thereafter, all animals developed a steep increase in fluid requirements. Median survival time was 290min with SB328437 and >300min after vehicle and INCB3284 treatment (p<0.05). Series 3: HS/FR as in series 2. INCB3284 at t = 60min and t = 200min reduced fluid requirements by 75% until t = 300min (p<0.05 vs. vehicle). Mortality was 70% with vehicle and zero with INCB3284 treatment (p<0.05). Series 4: INCB3284 and SB328437 did not affect survival time in a lethal HS model without FR. Our findings further support the assumption that blockade of the major CCL2 receptor CCR2 is a promising approach to improve FR after HS and document that the dosing of INCB3284 can be optimized.


Assuntos
Choque Hemorrágico , Ratos , Animais , Ratos Sprague-Dawley , Benzamidas , Hemorragia/complicações , Receptores CCR , Ressuscitação , Modelos Animais de Doenças
6.
Pharmacol Res ; 190: 106730, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36925091

RESUMO

We reported previously that α1-adrenoceptor (α1-AR) ligands inhibit chemokine receptor (CR) heteromerization partners of α1B/D-AR. The underlying mechanisms are unknown and in vivo evidence for such effects is missing. Utilizing CCR2 and α1B-AR as prototypical partners, we observed in recombinant systems and THP-1 cells that α1B-AR enhanced whereas its absence inhibited Gαi signaling of CCR2. Phenylephrine and phentolamine reduced the CCR2:α1B-AR heteromerization propensity and inhibited Gαi signaling of CCR2. Phenylephrine cross-recruited ß-arrestin-2 to CCR2, and reduced expression of α1B/D-AR, CR partners (CCR1/2, CXCR4) and corresponding heteromers. Phentolamine reduced CR:α1B/D-AR heteromers without affecting ß-arrestin-2 recruitment or receptor expression. Phenylephrine/phentolamine prevented leukocyte infiltration mediated via CR heteromerization partners in a murine air pouch model. Our findings document that α1-AR ligands inhibit leukocyte migration mediated by CR heteromerization partners in vivo and suggest interference with α1B-AR:CR heteromerization as a mechanism by which CR partners are inhibited. These findings provide new insights into the pharmacology of GPCR heteromers and indicate that an agonist and antagonist at one GPCR can act as antagonists at heteromerization partners of their target receptors.


Assuntos
Receptores Adrenérgicos alfa 1 , Receptores Adrenérgicos , Camundongos , Animais , Ligantes , Fentolamina , Fenilefrina/farmacologia , beta-Arrestina 2/metabolismo , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo
7.
FEBS Lett ; 596(20): 2706-2716, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35920096

RESUMO

Previously, we reported that chemokine (C-C motif) receptor 2 (CCR2) heteromerizes with α1B -adrenoceptor (α1B -AR) in leukocytes, through which α1B -AR controls CCR2. Whether such heteromers are expressed in human vascular smooth muscle cells (hVSMCs) is unknown. Bioluminescence resonance energy transfer confirmed formation of recombinant CCR2:α1b -AR heteromers. Proximity ligation assays detected CCR2:α1B -AR heteromers in hVSMCs and human mesenteric arteries. CCR2:α1B -AR heteromerization per se enhanced α1B -AR-mediated Gαq -coupling. Chemokine (C-C motif) ligand 2 (CCL2) binding to CCR2 inhibited Gαq activation via α1B -AR, cross-recruited ß-arrestin to and induced internalization of α1B -AR in recombinant systems and in hVSMCs. Our findings suggest that CCR2 within CCR2:α1B -AR heteromers biases α1B -AR signaling and provide a mechanism for previous observations suggesting a role for CCL2/CCR2 in the regulation of cardiovascular function.


Assuntos
Quimiocina CCL2 , Receptores Adrenérgicos alfa 1 , Humanos , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , beta-Arrestinas/metabolismo , Quimiocinas/metabolismo , Viés
8.
Crit Care Explor ; 4(5): e0701, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35620770

RESUMO

Clinical correlations suggest that systemic chemokine (C-C motif) ligand (CCL) 2 release may contribute to blood pressure regulation and the development of hemodynamic instability during the early inflammatory response to traumatic-hemorrhagic shock. Thus, we investigated whether blockade of the principal CCL2 receptor chemokine (C-C motif) receptor (CCR) 2 affects blood pressure in normal animals, and hemodynamics and resuscitation fluid requirements in hemorrhagic shock models. DESIGN: Randomized prospective treatment study. SETTING: University laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: First, treatment of healthy anesthetized rats with increasing doses of INCB3284 or vehicle. Second, rats were hemorrhaged for 30 minutes, followed by treatment with the CCR2 antagonist INCB3284 (1.1 and 5.5 µmol/kg), the CCR5 antagonist Maraviroc (=control, 5.5 µmol/kg) or vehicle, and subsequent fluid resuscitation to maintain blood pressure until t = 90 minutes. Third, treatment of rats with 5 µmol/kg INCB3284 or vehicle after hemorrhage and fluid resuscitation until t = 300 minutes. MEASUREMENTS AND MAIN RESULTS: INCB3284 did not affect intrinsic function of isolated rat resistance arteries in pressure myography experiments. Blood pressure in anesthetized vehicle-treated animals continuously decreased by 0.09 ± 0.01 mm Hg/min (p < 0.001) but remained constant after INCB3284 injections. Systemic concentrations of the CCR2 agonists CCL2, CCL5, and CCL11 increased during hemorrhage and fluid resuscitation. INCB3284 dose-dependently reduced fluid requirements by 58% ± 11% in short-term experiments, whereas Maraviroc and vehicle-treated animals were indistinguishable. When resuscitation was performed until t = 300 minutes, INCB3284 reduced fluid requirements by 62% ± 6%, prevented from hemodynamic decompensation, reduced mortality from 50% with vehicle treatment to zero, and reduced overall tissue wet-weight/dry-weight ratios. CONCLUSIONS: Our findings suggest that CCR2 is involved in the regulation of normal cardiovascular function and during the cardiovascular stress response to hemorrhagic shock and fluid resuscitation. The present study identifies CCR2 as a drug target to reduce fluid requirements and to prevent death from hemodynamic decompensation during resuscitation from hemorrhagic shock.

9.
Proc Natl Acad Sci U S A ; 119(20): e2123511119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35537053

RESUMO

It is known that catecholamines regulate innate immune functions. The underlying mechanisms, however, are not well understood. Here we show that at least 20 members of the human chemokine receptor (CR) family heteromerize with one or more members of the α1-adrenergic receptor (AR) family in recombinant systems and that such heteromeric complexes are detectable in human monocytes and the monocytic leukemia cell line THP-1. Ligand binding to α1-ARs inhibited migration toward agonists of the CR heteromerization partners of α1B/D-ARs with high potency and 50 to 77% efficacy but did not affect migration induced by a noninteracting CR. Incomplete siRNA knockdown of α1B/D-ARs in THP-1 cells partially inhibited migration toward agonists of their CR heteromerization partners. Complete α1B-AR knockout via CRISPR-Cas9 gene editing in THP-1 cells (THP-1_ADRA1BKO) resulted in 82% reduction of α1D-AR expression and did not affect CR expression. Migration of THP-1_ADRA1BKO cells toward agonists of CR heteromerization partners of α1B/D-ARs was reduced by 82 to 95%. Our findings indicate that CR:α1B/D-AR heteromers are essential for normal function of CR heteromerization partners, provide a mechanism underlying neuroendocrine control of leukocyte trafficking, and offer opportunities to modulate leukocyte and/or cancer cell trafficking in disease processes.


Assuntos
Movimento Celular , Leucócitos , Receptores Adrenérgicos alfa 1 , Receptores CXCR4 , Membrana Celular/metabolismo , Humanos , Leucócitos/metabolismo , Neoplasias , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais
10.
PLoS One ; 16(6): e0253821, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34166476

RESUMO

Recently, we reported that the chemokine (C-X-C motif) receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) heteromerize with α1A/B/D-adrenoceptors (ARs) and arginine vasopressin receptor 1A (AVPR1A) in recombinant systems and in rodent and human vascular smooth muscle cells (hVSMCs). In these studies, we observed that heteromerization between two receptor partners may depend on the presence and the expression levels of other partnering receptors. To test this hypothesis and to gain initial insight into the formation of these receptor heteromers in native cells, we utilized proximity ligation assays in hVSMCs to visualize receptor-receptor proximity and systematically studied how manipulation of the expression levels of individual protomers affect heteromerization patterns among other interacting receptor partners. We confirmed subtype-specific heteromerization between endogenously expressed α1A/B/D-ARs and detected that AVPR1A also heteromerizes with α1A/B/D-ARs. siRNA knockdown of CXCR4 and of ACKR3 resulted in a significant re-arrangement of the heteromerization patterns among α1-AR subtypes. Similarly, siRNA knockdown of AVPR1A significantly increased heteromerization signals for seven of the ten receptor pairs between CXCR4, ACKR3, and α1A/B/D-ARs. Our findings suggest plasticity of seven transmembrane helix (7TM) receptor heteromerization in native cells and could be explained by a supramolecular organization of these receptors within dynamic clusters in the plasma membrane. Because we previously observed that recombinant CXCR4, ACKR3, α1a-AR and AVPR1A form hetero-oligomeric complexes composed of 2-4 different protomers, which show signaling properties distinct from individual protomers, re-arrangements of receptor heteromerization patterns in native cells may contribute to the phenomenon of context-dependent GPCR signaling. Furthermore, these findings advise caution in the interpretation of functional consequences after 7TM receptor knockdown in experimental models. Alterations of the heteromerization patterns among other receptor partners may alter physiological and pathological responses, in particular in more complex systems, such as studies on the function of isolated organs or in in vivo experiments.


Assuntos
Regulação da Expressão Gênica , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Multimerização Proteica , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Linhagem Celular , Humanos , Regiões Promotoras Genéticas , Estrutura Secundária de Proteína
11.
J Basic Clin Physiol Pharmacol ; 33(4): 519-523, 2021 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-34144642

RESUMO

OBJECTIVES: Chemokine receptor antagonists are being explored for their therapeutic potential in various disease processes. As the chemokine (C-C motif) receptor 2 (CCR2) antagonist RS504393 is known to compete with ligand binding to α1-adrenoceptors, we tested a panel of 10 CCR antagonists for interactions with α1-adrenoceptors to evaluate potential cardiovascular activities and side-effect profiles. METHODS: The PRESTO-Tango ß-arrestin recruitment assay was utilized to test whether the CCR antagonists interfere with α1b-AR activation upon stimulation with phenylephrine. Pressure myography with isolated rat resistance arteries was employed to assess their effects on phenylephrine-induced vasoconstriction. The following antagonists were tested: CCR1-BX471, BX513, BI639667; CCR2-RS504393, INCB3284; CCR3-SB328437; and CCR4-AZD2098, and C021; CCR5-Maraviroc; CCR10-BI6901. The pan-α1-adrenoceptor antagonist prazosin was used as control. RESULTS: Among the CCR antagonists tested, RS504393, BX513, and C021 inhibited phenylephrine-induced ß-arrestin recruitment to α1b-adrenoceptor and phenylephrine-induced vasoconstriction. While RS504393 functioned as a competitive α1-adrenoceptor blocker, BX513 and C021 functioned as noncompetitive α1-adrenoceptor antagonists in both assay systems. Furthermore, RS504393, BX513, and C021 dose-dependently dilated arteries that were fully preconstricted with phenylephrine. CONCLUSIONS: Our data suggest that CCR antagonists should be screened for cross-reactivity with α1-adrenoceptors to exclude potential adverse cardiovascular effects when used as anti inflammatory drugs.


Assuntos
Antagonistas Adrenérgicos alfa , Prazosina , Antagonistas de Receptores Adrenérgicos alfa 1 , Animais , Fenilefrina , Ratos , Receptores Adrenérgicos alfa 1 , Receptores de Quimiocinas , beta-Arrestinas
12.
FEBS Lett ; 595(14): 1863-1875, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34032285

RESUMO

Although class A seven-transmembrane helix (7TM) receptor hetero-oligomers have been proposed, information on the assembly and function of such higher-order hetero-oligomers is not available. Utilizing bioluminescence resonance energy transfer (BRET), bimolecular luminescence/fluorescence complementation (BiLC/BiFC), and BiLC/BiFC BRET in HEK293T cells, we provide evidence that chemokine (C-X-C motif) receptor 4, atypical chemokine receptor 3, α1a -adrenoceptor, and arginine vasopressin receptor 1A form hetero-oligomers composed of 2-4 different protomers. We show that hetero-oligomerization per se and ligand binding to individual protomers regulate agonist-induced coupling to the signaling transducers of interacting receptor partners. Our findings support the concept that receptor hetero-oligomers form supramolecular machineries with molecular signaling properties distinct from the individual protomers. These findings provide a mechanism for the phenomenon of context-dependent receptor function.


Assuntos
Quimiocina CXCL12/metabolismo , Receptores Adrenérgicos alfa 1/química , Receptores CXCR4/química , Receptores CXCR/química , Receptores de Vasopressinas/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/farmacologia , Transferência Ressonante de Energia de Fluorescência , Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Cinética , Luciferases/genética , Luciferases/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
J Biol Chem ; 295(44): 14893-14905, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-32839271

RESUMO

The chemokine receptor CXCR4, a G protein-coupled receptor (GPCR) capable of heteromerizing with other GPCRs, is involved in many processes, including immune responses, hematopoiesis, and organogenesis. Evidence suggests that CXCR4 activation reduces thrombin/protease-activated receptor 1 (PAR1)-induced impairment of endothelial barrier function. However, the mechanisms underlying cross-talk between CXCR4 and PAR1 are not well-understood. Using intermolecular bioluminescence resonance energy transfer and proximity ligation assays, we found that CXCR4 heteromerizes with PAR1 in the HEK293T expression system and in human primary pulmonary endothelial cells (hPPECs). A peptide analog of transmembrane domain 2 (TM2) of CXCR4 interfered with PAR1:CXCR4 heteromerization. In HTLA cells, the presence of CXCR4 reduced the efficacy of thrombin to induce ß-arrestin-2 recruitment to recombinant PAR1 and enhanced thrombin-induced Ca2+ mobilization. Whereas thrombin-induced extracellular signal-regulated protein kinase 1/2 (ERK1/2) phosphorylation occurred more transiently in the presence of CXCR4, peak ERK1/2 phosphorylation was increased when compared with HTLA cells expressing PAR1 alone. CXCR4-associated effects on thrombin-induced ß-arrestin-2 recruitment to and signaling of PAR1 could be reversed by TM2. In hPPECs, TM2 inhibited thrombin-induced ERK1/2 phosphorylation and activation of Ras homolog gene family member A. CXCR4 siRNA knockdown inhibited thrombin-induced ERK1/2 phosphorylation. Whereas thrombin stimulation reduced surface expression of PAR1, CXCR4, and PAR1:CXCR4 heteromers, chemokine (CXC motif) ligand 12 stimulation reduced surface expression of CXCR4 and PAR1:CXCR4 heteromers, but not of PAR1. Finally, TM2 dose-dependently inhibited thrombin-induced impairment of hPPEC monolayer permeability. Our findings suggest that CXCR4:PAR1 heteromerization enhances thrombin-induced G protein signaling of PAR1 and PAR1-mediated endothelial barrier disruption.


Assuntos
Receptor PAR-1/metabolismo , Receptores CXCR4/metabolismo , Trombina/metabolismo , Biopolímeros/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Transferência de Energia , Células HEK293 , Humanos , Pulmão/citologia , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases , Fosforilação
14.
Sci Rep ; 10(1): 11359, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647374

RESUMO

We compared therapeutic properties of natural and engineered chemokine (C-X-C motif) receptor 4 (CXCR4) agonists in a rat acute respiratory distress syndrome (ARDS) model utilizing the PaO2/FiO2-ratio as a clinically relevant primary outcome criterion. Ventilated rats underwent unilateral lung ischemia from t = 0-70 min plus hemorrhage to a mean arterial blood pressure (MAP) of 30 mmHg from t = 40-70 min, followed by reperfusion/fluid resuscitation until t = 300 min. Natural CXCR4 agonists (CXCL12, ubiquitin) and engineered CXCL12 variants (CXCL121, CXCL22, CXCL12K27A/R41A/R47A, CXCL12 (3-68)) were administered within 5 min of fluid resuscitation. Animals treated with vehicle or CXCL12 (3-68) reached criteria for mild and moderate ARDS between t = 90-120 min and t = 120-180 min, respectively, and remained in moderate ARDS until t = 300 min. Ubiquitin, CXCL12, CXCL121 and CXCL122 prevented ARDS development. Potencies of CXCL12/CXCL121/CXCL122 were higher than the potency of ubiquitin. CXCL12K27A/R41A/R47A was inefficacious. CXCL121 > CXCL12 stabilized MAP and reduced fluid requirements. CXCR4 agonists at doses that preserved lung function reduced histological injury of the post-ischemic lung and reduced mortality from 55 to 9%. Our findings suggest that CXCR4 protein agonists prevent development of ARDS and reduce mortality in a rat model, and that development of new engineered protein therapeutics with improved pharmacological properties for ARDS is possible.


Assuntos
Receptores CXCR4/agonistas , Traumatismo por Reperfusão/prevenção & controle , Síndrome do Desconforto Respiratório/prevenção & controle , Ressuscitação/métodos , Choque Hemorrágico/terapia , Ferimentos e Lesões/terapia , Animais , Quimiocina CXCL12/administração & dosagem , Quimiocina CXCL12/genética , Modelos Animais de Doenças , Hidratação/métodos , Humanos , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Engenharia de Proteínas , Ratos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/mortalidade , Traumatismo por Reperfusão/patologia , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/mortalidade , Síndrome do Desconforto Respiratório/patologia , Choque Hemorrágico/etiologia , Choque Hemorrágico/mortalidade , Choque Hemorrágico/patologia , Toracotomia/efeitos adversos , Ubiquitina/administração & dosagem , Ferimentos e Lesões/complicações , Ferimentos e Lesões/mortalidade
15.
J Phys Condens Matter ; 32(35): 355404, 2020 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-32344387

RESUMO

Floquet Majorana edge modes capture the topological features of periodically driven p-wave superconductors. We present a Kitaev chain with multiple time periodic driving terms. Our results demonstrate how multiple driving will affect Floquet bands in frequency space, leading to more robust Floquet Majorana edge modes against driving frequency ω in comparison with the single driving scenario. Meanwhile, we have proposed how to predict Majorana edge modes via the Zak phase of Floquet bands. Besides, in contrast to the cases with single driving term, where the constant phase can be gauged out by properly choosing the initial time, we have shown the relative phase between multiple driving can not be gauged out and will play a dominant role in deciding topological phase transitions. For the sake of completeness, we also investigate the high frequency limit. Analytical results on effective Hamiltonian can be obtained via Magnus expansion and relative phase induced topological transitions can be shown explicitly.

16.
Biochem Biophys Res Commun ; 528(2): 368-375, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32085899

RESUMO

Recently, we reported that chemokine (C-X-C motif) receptor 4 (CXCR4) heteromerizes with α1-adrenergic receptors (AR) on the cell surface of vascular smooth muscle cells, through which the receptors cross-talk. Direct biophysical evidence for CXCR4:α1-AR heteromers, however, is lacking. Here we utilized bimolecular luminescence/fluorescence complementation (BiLC/BiFC) combined with intermolecular bioluminescence resonance energy transfer (BRET) assays in HEK293T cells to evaluate CXCR4:α1a/b/d-AR heteromerization. Atypical chemokine receptor 3 (ACKR3) and metabotropic glutamate receptor 1 (mGlu1R) were utilized as controls. BRET between CXCR4-RLuc (Renilla reniformis) and enhanced yellow fluorescent protein (EYFP)-tagged ACKR3 or α1a/b/d-ARs fulfilled criteria for constitutive heteromerization. BRET between CXCR4-RLuc and EYFP or mGlu1R-EYFP were nonspecific. BRET50 for CXCR4:ACKR3 and CXCR4:α1a/b/d-AR heteromers were comparable. Stimulation of cells with phenylephrine increased BRETmax of CXCR4:α1a/b/d-AR heteromers without affecting BRET50; stimulation with CXCL12 reduced BRETmax of CXCR4:α1a-AR heteromers, but did not affect BRET50 or BRETmax/50 for CXCR4:α1b/d-AR. A peptide analogue of transmembrane domain (TM) 2 of CXCR4 reduced BRETmax of CXCR4:α1a/b/d-AR heteromers and increased BRET50 of CXCR4:α1a/b-AR interactions. A TM4 analogue of CXCR4 did not alter BRET. We observed CXCR4, α1a-AR and mGlu1R homodimerization by BiFC/BiLC, and heteromerization of homodimeric CXCR4 with proto- and homodimeric α1a-AR by BiFC/BiLC BRET. BiFC/BiLC BRET for interactions between homodimeric CXCR4 and homodimeric mGlu1R was nonspecific. Our findings suggest that the heteromerization affinity of CXCR4 for ACKR3 and α1-ARs is comparable, provide evidence for conformational changes of the receptor complexes upon agonist binding and support the concept that proto- and oligomeric CXCR4 and α1-ARs constitutively form higher-order hetero-oligomeric receptor clusters.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Multimerização Proteica , Receptores Adrenérgicos alfa 1/metabolismo , Receptores CXCR4/metabolismo , Quimiocina CXCL12/farmacologia , Células HEK293 , Humanos , Peptídeos/farmacologia , Fenilefrina/farmacologia , Ligação Proteica , Receptores CXCR4/química
17.
RSC Adv ; 10(10): 5516-5524, 2020 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-35497417

RESUMO

Three commercial Ni-Al alloys formed by a vacuum atomization method (NAV), atmospheric atomization method (NAA) and high-temperature melting method (NAH) were leached by 10 wt% NaOH solution to prepare three RANEY®-Ni catalysts (RNAV, RNAA and RNAH, correspondingly). The effects of a forming process on the structure of Ni-Al alloys and the corresponding RANEY®-Ni catalysts were investigated via XRD, XPS, SEM, TEM, NH3-TPD, N2 adsorption-desorption and EDX-mapping studies. Also, the as-prepared RANEY®-Ni catalysts were evaluated via the hydrogenation of 1,4-butenediol (BED) to produce 1,4-butanediol (BDO). The results showed that the specific surface areas and surface morphologies of the Ni-Al alloys present significant differences. Meanwhile, the RNAA sample presented a comparatively regular morphology, similar to a small piece of sugar cane. The weak and medium acid peak areas of the RNAA catalyst were lower than those of the other samples. RNAV showed higher weak and medium acid peak areas, demonstrating the higher number of acid centers on the surface of the catalyst. The surface of the RNAA catalyst obtained from NAA contained more active component-Ni, about 90 wt% on the surface, and the specific surface area of the sample was 75 times that of its precursor Ni-Al alloy powder (NAA). The evaluation results present that the RNAA catalyst shows better hydrogenation performance, with BED conversion of 100%, both BDO selectivity and yield of 46.11%.

18.
Sci Rep ; 9(1): 8653, 2019 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-31209229

RESUMO

We investigate the transport problem that a spinful matter wave is incident on a strong localized spin-orbit-coupled Bose-Einstein condensate in optical lattices, where the localization is admitted by atom interaction only existing at one particular site, and the spin-orbit coupling arouse spatial rotation of the spin texture. We find that tuning the spin orientation of the localized Bose-Einstein condensate can lead to spin-nonreciprocal/spin-reciprocal transport, meaning the transport properties are dependent on/independent of the spin orientation of incident waves. In the former case, we obtain the conditions to achieve transparency, beam-splitting, and blockade of the incident wave with a given spin orientation, and furthermore the ones to perfectly isolate incident waves of different spin orientation, while in the latter, we obtain the condition to maximize the conversion of different spin states. The result may be useful to develop a novel spinful matter wave valve that integrates spin switcher, beam-splitter, isolator, and converter. The method can also be applied to other real systems, e.g., realizing perfect isolation of spin states in magnetism, which is otherwise rather difficult.

19.
Phys Rev E ; 99(4-1): 042102, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31108708

RESUMO

Thermal rectification and heat amplification are investigated in a nonequilibrium V-type three-level system with quantum interference. By applying the Redfield master equation combined with full counting statistics, we analyze the steady-state heat transport. The noise-induced interference is found to be able to rectify the heat current, which paves a new way to design quantum thermal rectifier. Within the three-reservoir setup, the heat amplification is clearly identified far from equilibrium, which is in absence of the negative differential thermal conductance.

20.
Sci Signal ; 11(552)2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30327409

RESUMO

Repeated dosing of drugs targeting G protein-coupled receptors can stimulate antagonist tolerance, which reduces their efficacy; thus, strategies to avoid tolerance are needed. The efficacy of AMD3100, a competitive antagonist of the chemokine receptor CXCR4 that mobilizes leukemic blasts from the bone marrow into the blood to sensitize them to chemotherapy, is reduced after prolonged treatment. Tolerance to AMD3100 increases the abundance of CXCR4 on the surface of leukemic blasts, which promotes their rehoming to the bone marrow. AMD3100 inhibits both G protein signaling by CXCR4 and ß-arrestin1/2-dependent receptor endocytosis. We demonstrated that biased antagonists of G protein-dependent chemotaxis but not ß-arrestin1/2 recruitment and subsequent receptor endocytosis avoided tolerance. The peptide antagonist X4-2-6, which is derived from transmembrane helix 2 and extracellular loop 1 of CXCR4, limited chemotaxis and signaling but did not promote CXCR4 accumulation on the cell surface or cause tolerance. The activity of X4-2-6 was due to its distinct mechanism of inhibition of CXCR4. The peptide formed a ternary complex with the receptor and its ligand, the chemokine CXCL12. Within this complex, X4-2-6 released the portion of CXCL12 critical for receptor-mediated activation of G proteins but enabled the rest of the chemokine to recruit ß-arrestins to the receptor. In contrast, AMD3100 displaced all components of the chemokine responsible for CXCR4 activation. We further identified a small molecule with similar biased antagonist properties to those of X4-2-6, which may provide a viable alternative to patients when antagonist tolerance prevents drugs from reaching efficacy.


Assuntos
Tolerância a Medicamentos , Proteínas de Ligação ao GTP/antagonistas & inibidores , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/química , Transdução de Sinais , Animais , Benzilaminas , Células CHO , Quimiocina CXCL12/metabolismo , Quimiotaxia , Cricetinae , Cricetulus , Ciclamos , Endocitose , Fibroblastos/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Humanos , Células Jurkat , Ligantes , Camundongos , Fosforilação , Domínios Proteicos , Células THP-1 , beta-Arrestina 1/metabolismo , beta-Arrestina 2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA