Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Cell ; 187(6): 1460-1475.e20, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38428423

RESUMEN

Apelin is a key hormone in cardiovascular homeostasis that activates the apelin receptor (APLNR), which is regarded as a promising therapeutic target for cardiovascular disease. However, adverse effects through the ß-arrestin pathway limit its pharmacological use. Here, we report cryoelectron microscopy (cryo-EM) structures of APLNR-Gi1 complexes bound to three agonists with divergent signaling profiles. Combined with functional assays, we have identified "twin hotspots" in APLNR as key determinants for signaling bias, guiding the rational design of two exclusive G-protein-biased agonists WN353 and WN561. Cryo-EM structures of WN353- and WN561-stimulated APLNR-G protein complexes further confirm that the designed ligands adopt the desired poses. Pathophysiological experiments have provided evidence that WN561 demonstrates superior therapeutic effects against cardiac hypertrophy and reduced adverse effects compared with the established APLNR agonists. In summary, our designed APLNR modulator may facilitate the development of next-generation cardiovascular medications.


Asunto(s)
Receptores de Apelina , Fármacos Cardiovasculares , Diseño de Fármacos , Receptores de Apelina/agonistas , Receptores de Apelina/química , Receptores de Apelina/ultraestructura , Microscopía por Crioelectrón , Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Humanos , Fármacos Cardiovasculares/química
2.
Cell ; 186(26): 5784-5797.e17, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38101408

RESUMEN

Cannabis activates the cannabinoid receptor 1 (CB1), which elicits analgesic and emotion regulation benefits, along with adverse effects, via Gi and ß-arrestin signaling pathways. However, the lack of understanding of the mechanism of ß-arrestin-1 (ßarr1) coupling and signaling bias has hindered drug development targeting CB1. Here, we present the high-resolution cryo-electron microscopy structure of CB1-ßarr1 complex bound to the synthetic cannabinoid MDMB-Fubinaca (FUB), revealing notable differences in the transducer pocket and ligand-binding site compared with the Gi protein complex. ßarr1 occupies a wider transducer pocket promoting substantial outward movement of the TM6 and distinctive twin toggle switch rearrangements, whereas FUB adopts a different pose, inserting more deeply than the Gi-coupled state, suggesting the allosteric correlation between the orthosteric binding pocket and the partner protein site. Taken together, our findings unravel the molecular mechanism of signaling bias toward CB1, facilitating the development of CB1 agonists.


Asunto(s)
Arrestina , Receptor Cannabinoide CB1 , Transducción de Señal , Arrestina/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo , Microscopía por Crioelectrón , Receptor Cannabinoide CB1/metabolismo , Humanos , Animales , Línea Celular
3.
Cell ; 184(4): 931-942.e18, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571431

RESUMEN

The D1- and D2-dopamine receptors (D1R and D2R), which signal through Gs and Gi, respectively, represent the principal stimulatory and inhibitory dopamine receptors in the central nervous system. D1R and D2R also represent the main therapeutic targets for Parkinson's disease, schizophrenia, and many other neuropsychiatric disorders, and insight into their signaling is essential for understanding both therapeutic and side effects of dopaminergic drugs. Here, we report four cryoelectron microscopy (cryo-EM) structures of D1R-Gs and D2R-Gi signaling complexes with selective and non-selective dopamine agonists, including two currently used anti-Parkinson's disease drugs, apomorphine and bromocriptine. These structures, together with mutagenesis studies, reveal the conserved binding mode of dopamine agonists, the unique pocket topology underlying ligand selectivity, the conformational changes in receptor activation, and potential structural determinants for G protein-coupling selectivity. These results provide both a molecular understanding of dopamine signaling and multiple structural templates for drug design targeting the dopaminergic system.


Asunto(s)
Receptores de Dopamina D1/química , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/química , Receptores de Dopamina D2/metabolismo , Transducción de Señal , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Secuencia de Aminoácidos , Secuencia Conservada , Microscopía por Crioelectrón , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Dopamina D1/ultraestructura , Receptores de Dopamina D2/ultraestructura , Homología Estructural de Proteína
4.
Mol Cell ; 84(3): 570-583.e7, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38215752

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) are evolutionarily ancient receptors involved in a variety of physiological and pathophysiological processes. Modulators of aGPCR, particularly antagonists, hold therapeutic promise for diseases like cancer and immune and neurological disorders. Hindered by the inactive state structural information, our understanding of antagonist development and aGPCR activation faces challenges. Here, we report the cryo-electron microscopy structures of human CD97, a prototypical aGPCR that plays crucial roles in immune system, in its inactive apo and G13-bound fully active states. Compared with other family GPCRs, CD97 adopts a compact inactive conformation with a constrained ligand pocket. Activation induces significant conformational changes for both extracellular and intracellular sides, creating larger cavities for Stachel sequence binding and G13 engagement. Integrated with functional and metadynamics analyses, our study provides significant mechanistic insights into the activation and signaling of aGPCRs, paving the way for future drug discovery efforts.


Asunto(s)
Antígenos CD , Receptores Acoplados a Proteínas G , Transducción de Señal , Humanos , Adhesión Celular , Microscopía por Crioelectrón , Complejo GPIb-IX de Glicoproteína Plaquetaria , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Antígenos CD/química , Antígenos CD/metabolismo
5.
Mol Cell ; 82(14): 2681-2695.e6, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35714614

RESUMEN

Serotonin (or 5-hydroxytryptamine, 5-HT) is an important neurotransmitter that activates 12 different G protein-coupled receptors (GPCRs) through selective coupling of Gs, Gi, or Gq proteins. The structural basis for G protein subtype selectivity by these GPCRs remains elusive. Here, we report the structures of the serotonin receptors 5-HT4, 5-HT6, and 5-HT7 with Gs, and 5-HT4 with Gi1. The structures reveal that transmembrane helices TM5 and TM6 alternate lengths as a macro-switch to determine receptor's selectivity for Gs and Gi, respectively. We find that the macro-switch by the TM5-TM6 length is shared by class A GPCR-G protein structures. Furthermore, we discover specific residues within TM5 and TM6 that function as micro-switches to form specific interactions with Gs or Gi. Together, these results present a common mechanism of Gs versus Gi protein coupling selectivity or promiscuity by class A GPCRs and extend the basis of ligand recognition at serotonin receptors.


Asunto(s)
Receptores Acoplados a Proteínas G , Serotonina , Proteínas de Unión al GTP/metabolismo , Ligandos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo
6.
Mol Cell ; 81(6): 1147-1159.e4, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33548201

RESUMEN

The dopamine system, including five dopamine receptors (D1R-D5R), plays essential roles in the central nervous system (CNS), and ligands that activate dopamine receptors have been used to treat many neuropsychiatric disorders. Here, we report two cryo-EM structures of human D3R in complex with an inhibitory G protein and bound to the D3R-selective agonists PD128907 and pramipexole, the latter of which is used to treat patients with Parkinson's disease. The structures reveal agonist binding modes distinct from the antagonist-bound D3R structure and conformational signatures for ligand-induced receptor activation. Mutagenesis and homology modeling illuminate determinants of ligand specificity across dopamine receptors and the mechanisms for Gi protein coupling. Collectively our work reveals the basis of agonist binding and ligand-induced receptor activation and provides structural templates for designing specific ligands to treat CNS diseases targeting the dopaminergic system.


Asunto(s)
Microscopía por Crioelectrón , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Modelos Moleculares , Complejos Multiproteicos/ultraestructura , Receptores de Dopamina D3/química , Benzopiranos/química , Células HEK293 , Humanos , Complejos Multiproteicos/química , Oxazinas/química , Pramipexol/química , Dominios Proteicos , Relación Estructura-Actividad
7.
Mol Cell ; 77(3): 669-680.e4, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32004470

RESUMEN

Corticotropin-releasing factor (CRF) and the three related peptides urocortins 1-3 (UCN1-UCN3) are endocrine hormones that control the stress responses by activating CRF1R and CRF2R, two members of class B G-protein-coupled receptors (GPCRs). Here, we present two cryoelectron microscopy (cryo-EM) structures of UCN1-bound CRF1R and CRF2R with the stimulatory G protein. In both structures, UCN1 adopts a single straight helix with its N terminus dipped into the receptor transmembrane bundle. Although the peptide-binding residues in CRF1R and CRF2R are different from other members of class B GPCRs, the residues involved in receptor activation and G protein coupling are conserved. In addition, both structures reveal bound cholesterol molecules to the receptor transmembrane helices. Our structures define the basis of ligand-binding specificity in the CRF receptor-hormone system, establish a common mechanism of class B GPCR activation and G protein coupling, and provide a paradigm for studying membrane protein-lipid interactions for class B GPCRs.


Asunto(s)
Receptores de Hormona Liberadora de Corticotropina/ultraestructura , Secuencia de Aminoácidos , Sitios de Unión , Hormona Liberadora de Corticotropina , Microscopía por Crioelectrón/métodos , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Péptidos/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Urocortinas/metabolismo
8.
Nature ; 594(7864): 594-598, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33911284

RESUMEN

G-protein-coupled receptors (GPCRs) have central roles in intercellular communication1,2. Structural studies have revealed how GPCRs can activate G proteins. However, whether this mechanism is conserved among all classes of GPCR remains unknown. Here we report the structure of the class-C heterodimeric GABAB receptor, which is activated by the inhibitory transmitter GABA, in its active form complexed with Gi1 protein. We found that a single G protein interacts with the GB2 subunit of the GABAB receptor at a site that mainly involves intracellular loop 2 on the side of the transmembrane domain. This is in contrast to the G protein binding in a central cavity, as has been observed with other classes of GPCR. This binding mode results from the active form of the transmembrane domain of this GABAB receptor being different from that of other GPCRs, as it shows no outside movement of transmembrane helix 6. Our work also provides details of the inter- and intra-subunit changes that link agonist binding to G-protein activation in this heterodimeric complex.


Asunto(s)
Proteínas de Unión al GTP/química , Receptores de GABA-B/química , Microscopía por Crioelectrón , Humanos , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Estructura Terciaria de Proteína
9.
Nature ; 592(7854): 469-473, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33762731

RESUMEN

Serotonin, or 5-hydroxytryptamine (5-HT), is an important neurotransmitter1,2 that activates the largest subtype family of G-protein-coupled receptors3. Drugs that target 5-HT1A, 5-HT1D, 5-HT1E and other 5-HT receptors are used to treat numerous disorders4. 5-HT receptors have high levels of basal activity and are subject to regulation by lipids, but the structural basis for the lipid regulation and basal activation of these receptors and the pan-agonism of 5-HT remains unclear. Here we report five structures of 5-HT receptor-G-protein complexes: 5-HT1A in the apo state, bound to 5-HT or bound to the antipsychotic drug aripiprazole; 5-HT1D bound to 5-HT; and 5-HT1E in complex with a 5-HT1E- and 5-HT1F-selective agonist, BRL-54443. Notably, the phospholipid phosphatidylinositol 4-phosphate is present at the G-protein-5-HT1A interface, and is able to increase 5-HT1A-mediated G-protein activity. The receptor transmembrane domain is surrounded by cholesterol molecules-particularly in the case of 5-HT1A, in which cholesterol molecules are directly involved in shaping the ligand-binding pocket that determines the specificity for aripiprazol. Within the ligand-binding pocket of apo-5-HT1A are structured water molecules that mimic 5-HT to activate the receptor. Together, our results address a long-standing question of how lipids and water molecules regulate G-protein-coupled receptors, reveal how 5-HT acts as a pan-agonist, and identify the determinants of drug recognition in 5-HT receptors.


Asunto(s)
Microscopía por Crioelectrón , Ligandos , Lípidos , Receptores de Serotonina 5-HT1/metabolismo , Receptores de Serotonina 5-HT1/ultraestructura , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Aripiprazol/metabolismo , Aripiprazol/farmacología , Sitios de Unión , Colesterol/farmacología , Proteínas de Unión al GTP Heterotriméricas/química , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas de Unión al GTP Heterotriméricas/ultraestructura , Humanos , Modelos Moleculares , Fosfatos de Fosfatidilinositol/química , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatos de Fosfatidilinositol/farmacología , Receptor de Serotonina 5-HT1A/química , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1A/ultraestructura , Receptores de Serotonina 5-HT1/química , Agonistas del Receptor de Serotonina 5-HT1/química , Agonistas del Receptor de Serotonina 5-HT1/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Agua/química
10.
Nature ; 589(7843): 620-626, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33408414

RESUMEN

Adhesion G-protein-coupled receptors (GPCRs) are a major family of GPCRs, but limited knowledge of their ligand regulation or structure is available1-3. Here we report that glucocorticoid stress hormones activate adhesion G-protein-coupled receptor G3 (ADGRG3; also known as GPR97)4-6, a prototypical adhesion GPCR. The cryo-electron microscopy structures of GPR97-Go complexes bound to the anti-inflammatory drug beclomethasone or the steroid hormone cortisol revealed that glucocorticoids bind to a pocket within the transmembrane domain. The steroidal core of glucocorticoids is packed against the 'toggle switch' residue W6.53, which senses the binding of a ligand and induces activation of the receptor. Active GPR97 uses a quaternary core and HLY motif to fasten the seven-transmembrane bundle and to mediate G protein coupling. The cytoplasmic side of GPR97 has an open cavity, where all three intracellular loops interact with the Go protein, contributing to the high basal activity of GRP97. Palmitoylation at the cytosolic tail of the Go protein was found to be essential for efficient engagement with GPR97 but is not observed in other solved GPCR complex structures. Our work provides a structural basis for ligand binding to the seven-transmembrane domain of an adhesion GPCR and subsequent G protein coupling.


Asunto(s)
Microscopía por Crioelectrón , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Glucocorticoides/química , Glucocorticoides/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/ultraestructura , Sitios de Unión , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/ultraestructura , Humanos , Ligandos , Lipoilación , Modelos Moleculares , Unión Proteica , Receptores Acoplados a Proteínas G/metabolismo
11.
Nature ; 587(7834): 499-504, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32698187

RESUMEN

The G-protein-coupled bile acid receptor (GPBAR) conveys the cross-membrane signalling of a vast variety of bile acids and is a signalling hub in the liver-bile acid-microbiota-metabolism axis1-3. Here we report the cryo-electron microscopy structures of GPBAR-Gs complexes stabilized by either the high-affinity P3954 or the semisynthesized bile acid derivative INT-7771,3 at 3 Å resolution. These structures revealed a large oval pocket that contains several polar groups positioned to accommodate the amphipathic cholic core of bile acids, a fingerprint of key residues to recognize diverse bile acids in the orthosteric site, a putative second bile acid-binding site with allosteric properties and structural features that contribute to bias properties. Moreover, GPBAR undertakes an atypical mode of activation and G protein coupling that features a different set of key residues connecting the ligand-binding pocket to the Gs-coupling site, and a specific interaction motif that is localized in intracellular loop 3. Overall, our study not only reveals unique structural features of GPBAR that are involved in bile acid recognition and allosteric effects, but also suggests the presence of distinct connecting mechanisms between the ligand-binding pocket and the G-protein-binding site in the G-protein-coupled receptor superfamily.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Microscopía por Crioelectrón , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/ultraestructura , Regulación Alostérica/efectos de los fármacos , Ácidos y Sales Biliares/química , Sitios de Unión/efectos de los fármacos , Ácidos Cólicos/química , Ácidos Cólicos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/ultraestructura , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Especificidad por Sustrato
12.
Proc Natl Acad Sci U S A ; 119(29): e2117054119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858343

RESUMEN

The G protein-coupled bile acid receptor (GPBAR) is the membrane receptor for bile acids and a driving force of the liver-bile acid-microbiota-organ axis to regulate metabolism and other pathophysiological processes. Although GPBAR is an important therapeutic target for a spectrum of metabolic and neurodegenerative diseases, its activation has also been found to be linked to carcinogenesis, leading to potential side effects. Here, via functional screening, we found that two specific GPBAR agonists, R399 and INT-777, demonstrated strikingly different regulatory effects on the growth and apoptosis of non-small cell lung cancer (NSCLC) cells both in vitro and in vivo. Further mechanistic investigation showed that R399-induced GPBAR activation displayed an obvious bias for ß-arrestin 1 signaling, thus promoting YAP signaling activation to stimulate cell proliferation. Conversely, INT-777 preferentially activated GPBAR-Gs signaling, thus inactivating YAP to inhibit cell proliferation and induce apoptosis. Phosphorylation of GPBAR by GRK2 at S310/S321/S323/S324 sites contributed to R399-induced GPBAR-ß-arrestin 1 association. The cryoelectron microscopy (cryo-EM) structure of the R399-bound GPBAR-Gs complex enabled us to identify key interaction residues and pivotal conformational changes in GPBAR responsible for the arrestin signaling bias and cancer cell proliferation. In summary, we demonstrate that different agonists can regulate distinct functions of cell growth and apoptosis through biased GPBAR signaling and control of YAP activity in a NSCLC cell model. The delineated mechanism and structural basis may facilitate the rational design of GPBAR-targeting drugs with both metabolic and anticancer benefits.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Proteínas de Ciclo Celular , Neoplasias Pulmonares , Receptores Acoplados a Proteínas G , Factores de Transcripción , Ácidos y Sales Biliares/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas de Ciclo Celular/metabolismo , Ácidos Cólicos/farmacología , Microscopía por Crioelectrón , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción/metabolismo , beta-Arrestina 1/metabolismo
13.
Med Res Rev ; 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39180380

RESUMEN

Chemotherapies are commonly used in cancer therapy, their applications are limited to low specificity, severe adverse reactions, and long-term medication-induced drug resistance. Poly(ADP-ribose) polymerase (PARP) inhibitors are a novel class of antitumor drugs developed to solve these intractable problems based on the mechanism of DNA damage repair, which have been widely applied in the treatment of ovarian cancer, breast cancer, and other cancers through inducing synthetic lethal effect and trapping PARP-DNA complex in BRCA gene mutated cancer cells. In recent years, PARP inhibitors have been widely used in combination with various first-line chemotherapy drugs, targeted drugs and immune checkpoint inhibitors to expand the scope of clinical application. However, the intricate mechanisms underlying the drug resistance to PARP inhibitors, including the restoration of homologous recombination, stabilization of DNA replication forks, overexpression of drug efflux protein, and epigenetic modifications pose great challenges and desirability in the development of novel PARP inhibitors. In this review, we will focus on the mechanism, structure-activity relationship, and multidrug resistance associated with the representative PARP inhibitors. Furthermore, we aim to provide insights into the development prospects and emerging trends to offer guidance for the clinical application and inspiration for the development of novel PARP inhibitors and degraders.

14.
Nat Chem Biol ; 18(3): 264-271, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34949837

RESUMEN

Biased signaling of G protein-coupled receptors describes an ability of different ligands that preferentially activate an alternative downstream signaling pathway. In this work, we identified and characterized different N-terminal truncations of endogenous chemokine CCL15 as balanced or biased agonists targeting CCR1, and presented three cryogenic-electron microscopy structures of the CCR1-Gi complex in the ligand-free form or bound to different CCL15 truncations with a resolution of 2.6-2.9 Å, illustrating the structural basis of natural biased signaling that initiates an inflammation response. Complemented with pharmacological and computational studies, these structures revealed it was the conformational change of Tyr291 (Y2917.43) in CCR1 that triggered its polar network rearrangement in the orthosteric binding pocket and allosterically regulated the activation of ß-arrestin signaling. Our structure of CCL15-bound CCR1 also exhibited a critical site for ligand binding distinct from many other chemokine-receptor complexes, providing new insights into the mode of chemokine recognition.


Asunto(s)
Proteínas de Unión al GTP , Receptores de Quimiocina , Quimiocinas/metabolismo , Quimiocinas/farmacología , Proteínas de Unión al GTP/metabolismo , Ligandos , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/metabolismo , beta-Arrestinas/metabolismo
15.
Environ Res ; 245: 118021, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38147917

RESUMEN

Saltwater intrusion in estuarine ecosystems alters microbial communities as well as biogeochemical cycling processes and has become a worldwide problem. However, the impact of salinity intrusion on the dynamics of nitrous oxide (N2O) and associated microbial community are understudied. Here, we conducted field microcosms in a tidal estuary during different months (December, April and August) using dialysis bags, and microbes inside the bags encountered a change in salinity in natural setting. We then compared N2O dynamics in the microcosms with that in natural water. Regardless of incubation environment, saltwater intrusion altered the dissolved N2O depending on the initial saturation rates of N2O. While the impact of saltwater intrusion on N2O dynamics was consistent across months, the dissolved N2O was higher in summer than in winter. The N-related microbial communities following saltwater intrusion were dominated by denitrifers, with fewer nitrifiers and bacterial taxa involved in dissimilatory nitrate reduction to ammonium. While denitrification was a significant driver of N2O dynamics in the studied estuary, nitrifier-involved denitrification contributed to the additional production of N2O, evidenced by the strong associations with amoA genes and the abundance of Nitrospira. Higher N2O concentrations in the field microcosms than in natural water limited N2O consumption in the former, given the lack of an association with nosZ gene abundance. The differences in the N2O dynamics observed between the microcosms and natural water could be that the latter comprised not only indigenous microbes but also those accompanied with saltwater intrusion, and that immigrants might be functionally rich individuals and able to perform N transformation in multiple pathways. Our work provides the first quantitative assessment of in situ N2O concentrations in an estuary subjected to a saltwater intrusion. The results highlight the importance of ecosystem size and microbial connectivity in the source-sink dynamics of N2O in changing environments.


Asunto(s)
Bacterias , Microbiota , Humanos , Bacterias/genética , Agua , Nitratos , Óxido Nitroso , Suelo
16.
Artículo en Inglés | MEDLINE | ID: mdl-38430180

RESUMEN

Context: Periprosthetic joint infections (PJIs) are a rare but highly destructive complication after total knee arthroplasty (TKA). Nursing plays an important role in preventing postoperative infections in patients, but different nursing modes have different rates of postoperative infections. Objective: The study intended to explore the effects of "encouragement, education, exercise, employment, and evaluation" (5E) rehabilitation nursing on the prevention of periprosthetic joint infections (PJIs) after TKA. Design: The research team conducted a randomized controlled trial. Setting: The study took place at the First People's Hospital of Huzhou in Huzhou, China. Participants: Participants were 80 TKA postoperative patients at the hospital between January 2023 and July 2023. Interventions: The research randomly divided participants into two groups: (1) the intervention group, the 5E group, with 40 participants and (2) the control group, with 40 participants. The control group received routine nursing, while the 5E group received 5E rehabilitation nursing. Outcome Measures: The research team examined: (1) the prosthesis' location; (2) wound healing; (3) score for knee joint function, using the Berg Balance Scale (BBS) and the Hospital Score for Special Surgery (HSS) of the knee joint; (4) postoperative level of inflammatory factors, using levels of C-reactive protein (CRP); (5) infection occurrence; (6) length of hospital stay; and (7) nursing satisfaction. Results: The prosthesis was well positioned in both groups. Compared to the control group, the 5E group's: (1) wound healing was significantly better (P < .001); (2) at 7 days after surgery, HSS score (P < .001) and BBS score (P < .001) were significantly higher; (3) C-reactive protein (CRP) levels were significantly lower (P < .001); (4) at 90 days after surgery, incidence of postoperative periprosthetic joint infection (PJI) was significantly lower (P < .001); (5) length of hospital stay was significantly shorter (P = .0013); and (7) nursing satisfaction was significantly higher (P = .0338). Conclusions: The 5E rehabilitation nursing for patients after TKA was helpful in promoting wound recovery, supporting the recovery of knee-joint function, reducing the incidence of PJIs, shortening the length of hospital stay, and improving patients' nursing satisfaction.

17.
Ecotoxicol Environ Saf ; 276: 116281, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38581907

RESUMEN

Bromophenols (BPs) are prominent environmental pollutants extensively utilized in aquaculture, pharmaceuticals, and chemical manufacturing. This study aims to identify UDP- glucuronosyltransferases (UGTs) isoforms involved in the metabolic elimination of BPs. Mono-glucuronides of BPs were detected in human liver microsomes (HLMs) incubated with the co-factor uridine-diphosphate glucuronic acid (UDPGA). The glucuronidation metabolism reactions catalyzed by HLMs followed Michaelis-Menten or substrate inhibition kinetics. Recombinant enzymes and inhibition experiments with chemical reagents were employed to phenotype the principal UGT isoforms participating in BP glucuronidation. UGT1A6 emerged as the major enzyme in the glucuronidation of 4-Bromophenol (4-BP), while UGT1A1, UGT1A6, and UGT1A8 were identified as the most essential isoforms for metabolizing 2,4-dibromophenol (2,4-DBP). UGT1A1, UGT1A8, and UGT2B4 were deemed the most critical isoforms in the catalysis of 2,4,6-tribromophenol (2,4,6-TBP) glucuronidation. Species differences were investigated using the liver microsomes of pig (PLM), rat (RLM), monkey (MyLM), and dog (DLM). Additionally, 2,4,6-TBP effects on the expression of UGT1A1 and UGT2B7 in HepG2 cells were evaluated. The results demonstrated potential induction of UGT1A1 and UGT2B7 upon exposure to 2,4,6-TBP at a concentration of 50 µM. Collectively, these findings contribute to elucidating the metabolic elimination and toxicity of BPs.


Asunto(s)
Glucurónidos , Glucuronosiltransferasa , Microsomas Hepáticos , Fenoles , Glucuronosiltransferasa/metabolismo , Humanos , Animales , Fenoles/toxicidad , Fenoles/metabolismo , Glucurónidos/metabolismo , Contaminantes Ambientales/toxicidad , Contaminantes Ambientales/metabolismo , Perros , Ratas , Isoenzimas/metabolismo , Especificidad de la Especie
18.
Environ Monit Assess ; 196(9): 818, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39150577

RESUMEN

Land use change stands as the primary factor influencing habitat quality (HQ). Clarifying the spatiotemporal change and the obstacle factors of the coupling relationship between HQ and urbanization level (UL) can provide imperative references for achieving sustainability in the Yellow River Basin (YRB). This study is based on the InVEST model, spatial autocorrelation, and obstacle factor analysis to measure the spatiotemporal dynamics and impediments of the coupling relationship between HQ and UL from 2000 to 2020 in the YRB. The findings were as follows: (1) From 2000 to 2020, the HQ showed a tendency of rise first and then fall, with the pattern of "High in the middle and west, low in the east"; (2) from 2000 to 2020, the UL had an upward trend, with the pattern of "Low in the west, high in the middle and east"; (3) the coupling and coordination level of HQ and UL in the YRB changed from extreme incoordination to verge of coordination, and it had a distribution pattern of "High in the east, low in the west", with the high-value area expanding to the east and the low-value area shrinking to the west. (4) Location condition, climate, proportion of construction land, vegetation index, and proportion of non-agricultural employment are the main obstacle factors that determined the coupling and coordination of the HQ and UL.


Asunto(s)
Ecosistema , Monitoreo del Ambiente , Ríos , Urbanización , China , Ríos/química , Conservación de los Recursos Naturales , Análisis Espacio-Temporal
19.
Med Res Rev ; 43(6): 2352-2391, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37211904

RESUMEN

The U.S. Food and Drug Administration has approved a total of 37 new drugs in 2022, which are composed of 20 chemical entities and 17 biologics. In particular, 20 chemical entities, including 17 small molecule drugs, 1 radiotherapy, and 2 diagnostic agents, provide privileged scaffolds, breakthrough clinical benefits, and a new mechanism of action for the discovery of more potent clinical candidates. The structure-based drug development with clear targets and fragment-based drug development with privileged scaffolds have always been the important modules in the field of drug discovery, which could easily bypass the patent protection and bring about improved biological activity. Therefore, we summarized the relevant valuable information about clinical application, mechanism of action, and chemical synthesis of 17 newly approved small molecule drugs in 2022. We hope this timely and comprehensive review could bring about creative and elegant inspiration on the synthetic methodologies and mechanism of action for the discovery of new drugs with novel chemical scaffolds and extended clinical indications.

20.
J Biomed Sci ; 30(1): 60, 2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37525190

RESUMEN

Dysregulation of various cells in the tumor microenvironment (TME) causes immunosuppressive functions and aggressive tumor growth. In combination with immune checkpoint blockade (ICB), epigenetic modification-targeted drugs are emerging as attractive cancer treatments. Lysine-specific demethylase 1 (LSD1) is a protein that modifies histone and non-histone proteins and is known to influence a wide variety of physiological processes. The dysfunction of LSD1 contributes to poor prognosis, poor patient survival, drug resistance, immunosuppression, etc., making it a potential epigenetic target for cancer therapy. This review examines how LSD1 modulates different cell behavior in TME and emphasizes the potential use of LSD1 inhibitors in combination with ICB therapy for future cancer research studies.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Histonas/metabolismo , Neoplasias/tratamiento farmacológico , Epigénesis Genética , Histona Demetilasas/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda