Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.208
Filtrar
1.
Bioorg Med Chem Lett ; 97: 129486, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37734424

RESUMO

Antibiotic resistance is a major threat to public health, and Gram-negative bacteria pose a particular challenge due to their combination of a low permeability cell envelope and efflux pumps. Our limited understanding of the chemical rules for overcoming these barriers represents a major obstacle in antibacterial drug discovery. Several recent efforts to address this problem have involved screening compound libraries for accumulation in bacteria in order to understand the structural properties required for Gram-negative permeability. Toward this end, we used cheminformatic analysis to design a library of sulfamidoadenosines (AMSN) having diverse substituents at the adenine C2 position. An efficient synthetic route was developed with installation of a uniform cross-coupling reagent set using Sonogashira and Suzuki reactions of a C2-iodide. The potential utility of these compounds was demonstrated by pilot analysis of selected analogues for accumulation in Escherichia coli.


Assuntos
Antibacterianos , Bactérias Gram-Negativas , Antibacterianos/química , Descoberta de Drogas , Escherichia coli , Permeabilidade/efeitos dos fármacos , Adenosina/química , Adenosina/farmacologia
2.
Front Immunol ; 14: 1110696, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36936939

RESUMO

Introduction: In an effort to minimize the usage of fishmeal in aquaculture, novel protein diets, including Tenebrio molitor, cottonseed protein concentrate, Clostridium autoethanogenum, and Chlorella vulgaris were evaluated for their potential to replace fishmeal. Nevertheless, comprehensive examinations on the gut health of aquatic animals under an alternate feeding strategy when fed novel protein diets are vacant. Methods: Five isonitrogenous and isolipidic diets containing various proteins were manufactured, with a diet consisting of whole fishmeal serving as the control and diets containing novel proteins serving as the experimental diets. Largemouth bass (Micropterus salmoides) with an initial body weight of 4.73 ± 0.04g employed as an experimental animal and given these five diets for the first 29 days followed by a fishmeal diet for the next 29 days. Results: The results of this study demonstrated that the growth performance of novel protein diets in the second stage was better than in the first stage, even though only the C. vulgaris diet increased antioxidant capacity and the cottonseed protein concentrate diet decreased it. Concerning the intestinal barriers, the C. autoethanogenum diet lowered intestinal permeability and plasma IL-1ß/TNF-α. In addition, the contents of intestinal immunological factors, namely LYS and sIgA-like, were greater in C. vulgaris than in fishmeal. From the data analysis of microbiome and metabolome, the levels of short chain fatty acids (SCFAs), anaerobic bacteria, Lactococcus, and Firmicutes were significantly higher in the C. autoethanogenum diet than in the whole fishmeal diet, while the abundance of Pseudomonas, aerobic bacteria, Streptococcus, and Proteobacteria was lowest. However, no extremely large differences in microbiota or short chain fatty acids were observed between the other novel protein diets and the whole fishmeal diet. In addition, the microbiota were strongly connected with intestinal SCFAs, lipase activity, and tight junctions, as shown by the Mantel test and Pearson's correlation. Discussion: Taken together, according to Z-score, the ranking of advantageous functions among these protein diets was C. autoethanogenum diet > C. vulgaris diet > whole fishmeal diet > cottonseed protein concentrate > T. molitor diet. This study provides comprehensive data illustrating a mixed blessing effect of novel protein diets on the gut health of juvenile largemouth bass under an alternate feeding strategy.


Assuntos
Ração Animal , Bass , Dieta , Intestinos , Bass/crescimento & desenvolvimento , Bass/imunologia , Bass/fisiologia , Multiômica , Intestinos/química , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/fisiologia , Proteínas de Peixes , Animais , Ração Animal/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Dieta/efeitos adversos , Dieta/métodos , Dieta/veterinária , Ácidos Graxos/análise , Óleo de Sementes de Algodão , Proteínas de Plantas , Chlorella vulgaris , Tenebrio , Insetos Comestíveis
3.
Alcohol ; 107: 136-143, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36150609

RESUMO

Alcohol use among older adults is on the rise. This increase is clinically relevant as older adults are at risk for increased morbidity and mortality from many alcohol-related chronic diseases compared to younger patients. However, little is known regarding the synergistic effects of alcohol and age. There are intriguing data suggesting that aging may lead to impaired intestinal barrier integrity and dysbiosis of the intestinal microbiome, which could increase susceptibility to alcohol's negative effects. To study the effects of alcohol in age we exposed aged and young mice to 3 days of moderate ethanol and evaluated changes in gut parameters. We found that these levels of drinking do not have obvious effects in young mice but cause significant alcohol-induced gut barrier dysfunction and expression of the pro-inflammatory cytokine TNFα in aged mice. Ethanol-induced downregulation of expression of the gut-protective antimicrobial peptides Defa-rs1, Reg3b, and Reg3g was observed in aged, but not young mice. Analysis of the fecal microbiome revealed age-associated shifts in microbial taxa, which correlated with intestinal and hepatic inflammatory gene expression. Taken together, these data demonstrate that age drives microbiome dysbiosis, while ethanol exposure in aged mice induces changes in the expression of antimicrobial genes important for separating these potentially damaging microbes from the intestinal lumen. These changes highlight potential mechanistic targets for prevention of the age-related exacerbation of effects of ethanol on the gut.


Assuntos
Disbiose , Etanol , Microbioma Gastrointestinal , Inflamação , Intestinos , Animais , Camundongos , Peptídeos Antimicrobianos/genética , Peptídeos Antimicrobianos/imunologia , Citocinas/imunologia , Disbiose/induzido quimicamente , Disbiose/genética , Disbiose/imunologia , Disbiose/microbiologia , Etanol/farmacologia , Etanol/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/microbiologia , Camundongos Endogâmicos C57BL , Permeabilidade/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , alfa-Defensinas/genética , alfa-Defensinas/imunologia
4.
Oxid Med Cell Longev ; 2022: 4834117, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251474

RESUMO

Overcoming blood-brain barrier (BBB) to improve brain bioavailability of therapeutic drug remains an ongoing concern. Prodrug is one of the most reliable approaches for delivering agents with low-level BBB permeability into the brain. The well-known antioxidant capacities of cysteine (Cys) and its vital role in glutathione (GSH) synthesis indicate that Cys-based prodrug could potentiate therapeutic drugs against oxidative stress-related neurodegenerative disorders. Moreover, prodrug with Cys moiety could be recognized by the excitatory amino acid transporter 3 (EAAT3) that is highly expressed at the BBB and transports drug into the brain. In this review, we summarized the strategies of crossing BBB, properties of EAAT3 and its natural substrates, Cys and its donors, and Cys donor-based brain-targeting prodrugs by referring to recent investigations. Moreover, the challenges that we are faced with and future research orientations were also addressed and proposed. It is hoped that present review will provide evidence for the pursuit of novel Cys donor-based brain-targeting prodrug.


Assuntos
Antioxidantes/metabolismo , Antioxidantes/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Cisteína/metabolismo , Cisteína/farmacologia , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Transportador 3 de Aminoácido Excitatório/metabolismo , Glutationa/metabolismo , Humanos , Permeabilidade/efeitos dos fármacos , Pró-Fármacos
5.
Oxid Med Cell Longev ; 2022: 9749461, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251482

RESUMO

Quercetin, a naturally occurring flavonoid, is mainly extracted from tea, onions, and apples. It has the underlying neuroprotective effect on experimental ischemic stroke. A systematic review and meta-analysis were used to assess quercetin's efficacy and possible mechanisms in treating focal cerebral ischemia. Compared with the control group, twelve studies reported a remarkable function of quercetin in improving the neurological function score (NFS) (P < 0.05), and twelve studies reported a significant effect on reducing infarct volume (P < 0.05). Moreover, two and three studies showed that quercetin could alleviate blood-brain barrier (BBB) permeability and brain water content, respectively. The mechanisms of quercetin against focal cerebral ischemia are diverse, involving antioxidation, antiapoptotic, anti-inflammation, and calcium overload reduction. On the whole, the present study suggested that quercetin can exert a protective effect on experimental ischemic stroke. Although the effect size may be overestimated because of the quality of studies and possible publication bias, these results indicated that quercetin might be a promising neuroprotective agent for human ischemic stroke. This study is registered with PROSPERO, number CRD 42021275656.


Assuntos
Infarto Cerebral/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Compostos Fitoquímicos/uso terapêutico , Fitoterapia/métodos , Extratos Vegetais/uso terapêutico , Quercetina/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Resultado do Tratamento
6.
Oxid Med Cell Longev ; 2022: 5044046, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222798

RESUMO

Cardiac lymphatic vessel growth (lymphangiogenesis) and integrity play an essential role in maintaining tissue fluid balance. Inhibition of lymphatic lymphangiogenesis is involved in cardiac edema and cardiac remodeling after ischemic injury or pressure overload. However, whether lymphatic vessel integrity is disrupted during angiotensin II- (Ang II-) induced cardiac remodeling remains to be investigated. In this study, cardiac remodeling models were established by Ang II (1000 ng/kg/min) in VEGFR-3 knockdown (Lyve-1Cre VEGFR-3f/-) and wild-type (VEGFR-3f/f) littermates. Our results indicated that Ang II infusion not only induced cardiac lymphangiogenesis and upregulation of VEGF-C and VEGFR-3 expression in the time-dependent manner but also enhanced proteasome activity, MKP5 and VE-cadherin degradation, p38 MAPK activation, and lymphatic vessel hyperpermeability. Moreover, VEGFR-3 knockdown significantly inhibited cardiac lymphangiogenesis in mice, resulting in exacerbation of tissue edema, hypertrophy, fibrosis superoxide production, inflammation, and heart failure (HF). Conversely, administration of epoxomicin (a selective proteasome inhibitor) markedly mitigated Ang II-induced cardiac edema, remodeling, and dysfunction; upregulated MKP5 and VE-cadherin expression; inactivated p38 MAPK; and reduced lymphatic vessel hyperpermeability in WT mice, indicating that inhibition of proteasome activity is required to maintain lymphatic endothelial cell (LEC) integrity. Our results show that both cardiac lymphangiogenesis and lymphatic barrier hyperpermeability are implicated in Ang II-induced adaptive hypertrophic remodeling and dysfunction. Proteasome-mediated hyperpermeability of LEC junctions plays a predominant role in the development of cardiac remodeling. Selective stimulation of lymphangiogenesis or inhibition of proteasome activity may be a potential therapeutic option for treating hypertension-induced cardiac remodeling.


Assuntos
Angiotensina II/metabolismo , Cardiomegalia/metabolismo , Edema Cardíaco/metabolismo , Vasos Linfáticos/metabolismo , Angiotensina II/administração & dosagem , Animais , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Edema Cardíaco/tratamento farmacológico , Edema Cardíaco/patologia , Edema Cardíaco/fisiopatologia , Células Endoteliais/metabolismo , Linfangiogênese/efeitos dos fármacos , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Permeabilidade/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/deficiência , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
Front Immunol ; 13: 847861, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35185936

RESUMO

Gut barrier disruption is the initial pathogenesis of various diseases. We previously reported that dietary allicin improves tight junction proteins in the endoplasmic reticulum stressed jejunum. However, whether the allicin benefits the gut barrier within mycotoxin or endotoxin exposure is unknown. In the present study, IPEC-J2 cell monolayers within or without deoxynivalenol (DON) or lipopolysaccharide (LPS) challenges were employed to investigate the effects of allicin on intestinal barrier function and explore the potential mechanisms. Results clarified that allicin at 2 µg/mL increased the viability, whereas the allicin higher than 10 µg/mL lowered the viability of IPEC-J2 cells via inhibiting cell proliferation. Besides, allicin increased trans-epithelial electric resistance (TEER), decreased paracellular permeability, and enhanced ZO-1 integrity of the IPEC-J2 cell monolayers. Finally, allicin supplementation prevented the LPS-induced barrier damages via activating Nrf2/HO-1 pathway-dependent antioxidant system. In conclusion, the present study strongly confirmed allicin as an effective nutrient to improve intestinal barrier function and prevent bacterial endotoxin-induced barrier damages.


Assuntos
Dissulfetos/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Jejuno/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Permeabilidade/efeitos dos fármacos , Ácidos Sulfínicos/farmacologia , Animais , Linhagem Celular , Impedância Elétrica , Células Epiteliais/metabolismo , Heme Oxigenase-1/metabolismo , Jejuno/metabolismo , Jejuno/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Suínos , Proteínas de Junções Íntimas/metabolismo
8.
Mol Med Rep ; 25(4)2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35147204

RESUMO

Ischemia/reperfusion (I/R) injury is a serious clinical condition characterized by high morbidity and mortality rates. Donepezil plays a neuroprotective role in I/R­associated diseases. The aim of the present study was to investigate the role and the potential mechanism of action of donepezil in I/R­induced myocardial microvascular endothelial cell dysfunction. An I/R model was simulated using oxygen­glucose deprivation/reoxygenation (OGD/R) injury in human cardiac microvascular endothelial cells (CMECs). Cell viability and lactate dehydrogenase release were examined following treatment with donepezil. Commercial kits were used to evaluate cell apoptosis, cell permeability and caspase­3 activity. The expression levels of apoptosis­associated proteins, as well as proteins found in tight junctions or involved in the poly(ADP­ribose) polymerase 1 (PARP1)/NF­κB pathway, were measured using western blotting. These parameters were also examined following PARP1 overexpression. The results demonstrated that donepezil increased cell viability and reduced toxicity in OGD/R­treated CMECs. The apoptotic rate, caspase­3 activity and protein expression levels of Bax and cleaved caspase­3 were significantly reduced following donepezil treatment, which was accompanied by Bcl­2 upregulation. Moreover, cell permeability was notably reduced, coupled with a marked increase in the expression of tight junction­associated proteins. The expression levels of proteins related to PARP1/NF­κB signaling were significantly downregulated in CMECs following donepezil treatment. However, the protective effects of donepezil on OGD/R­induced CMEC injury were reversed following PARP1 overexpression. In conclusion, donepezil suppressed OGD/R­induced CMEC dysfunction via PARP1/NF­κB signaling. This finding provided insight into the mechanism underlying myocardial I/R injury.


Assuntos
Donepezila/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , NF-kappa B/metabolismo , Fármacos Neuroprotetores/farmacologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Adolescente , Adulto , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Glucose/metabolismo , Humanos , Masculino , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Oxigênio/metabolismo , Permeabilidade/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/genética , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem
9.
Biochem Pharmacol ; 197: 114931, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35085542

RESUMO

Mitochondrial K+ permeability regulates neuronal apoptosis, energy metabolism, autophagy, and protection against ischemia-reperfusion injury. Kv7.4 channels have been recently shown to regulate K+ permeability in cardiac mitochondria and exert cardioprotective effects. Here, the possible expression and functional role of Kv7.4 channels in regulating membrane potential, radical oxygen species (ROS) production, and Ca2+ uptake in neuronal mitochondria was investigated in both clonal (F11 cells) and native brain neurons. In coupled mitochondria isolated from F11 cells, K+-dependent changes of mitochondrial membrane potential (ΔΨ) were unaffected by the selective mitoBKCa channel blocker iberiotoxin and only partially inhibited by the mitoKATP blockers glyburide or ATP. Interestingly, K+-dependent ΔΨ decrease was significantly reduced by the Kv7 blocker XE991 and enhanced by the Kv7 activator retigabine. Among Kv7s, western blot experiments showed the expression of only Kv7.4 subunits in F11 mitochondrial fractions; immunocytochemistry experiments showed a strong overlap between the Kv7.4 fluorescent signal and that of the mitochondrial marker Mitotracker. Silencing of Kv7.4 expression significantly suppressed retigabine-dependent decrease in ΔΨ in intact F11 cells. Expression of Kv7.4 subunits was also detected by western blot in isolated mitochondria from total mouse brain and by immunofluorescence in mouse primary cortical neurons. Pharmacological experiments revealed a relevant functional role for Kv7.4 channels in regulating membrane potential and Ca2+ uptake in isolated neuronal mitochondria, as well as ΔΨ and ROS production in intact cortical neurons. In conclusion, these findings provide the first experimental evidence for the expression of Kv7.4 channels and their contribution in regulating K+ permeability of neuronal mitochondria.


Assuntos
Canais de Potássio KCNQ/biossíntese , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Potássio/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Feminino , Glibureto/farmacologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Gravidez
10.
Biochim Biophys Acta Biomembr ; 1864(4): 183866, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35007517

RESUMO

In light of an increasing number of antibiotic-resistant bacterial strains, it is essential to understand an action imposed by various antimicrobial agents on bacteria at the molecular level. One of the leading mechanisms of killing bacteria is related to the alteration of their plasmatic membrane. We study bio-inspired peptides originating from natural antimicrobial proteins colicins, which can disrupt membranes of bacterial cells. Namely, we focus on the α-helix H1 of colicin U, produced by bacterium Shigella boydii, and compare it with analogous peptides derived from two different colicins. To address the behavior of the peptides in biological membranes, we employ a combination of molecular simulations and experiments. We use molecular dynamics simulations to show that all three peptides are stable in model zwitterionic and negatively charged phospholipid membranes. At the molecular level, their embedment leads to the formation of membrane defects, membrane permeation for water, and, for negatively charged lipids, membrane poration. These effects are caused by the presence of polar moieties in the considered peptides. Importantly, simulations demonstrate that even monomeric H1 peptides can form toroidal pores. At the macroscopic level, we employ experimental co-sedimentation and fluorescence leakage assays. We show that the H1 peptide of colicin U incorporates into phospholipid vesicles and disrupts their membranes, causing leakage, in agreement with the molecular simulations. These insights obtained for model systems seem important for understanding the mechanisms of antimicrobial action of natural bacteriocins and for future exploration of small bio-inspired peptides able to disrupt bacterial membranes.


Assuntos
Colicinas/metabolismo , Fosfolipídeos/química , Lipossomas Unilamelares/metabolismo , Sequência de Aminoácidos , Colicinas/química , Colicinas/farmacologia , Simulação de Dinâmica Molecular , Permeabilidade/efeitos dos fármacos , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Conformação Proteica em alfa-Hélice , Shigella boydii/metabolismo , Lipossomas Unilamelares/química
11.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35054928

RESUMO

The brain capillary endothelium is highly regulatory, maintaining the chemical stability of the brain's microenvironment. The role of cytoskeletal proteins in tethering nanotubules (TENTs) during barrier-genesis was investigated using the established immortalized mouse brain endothelial cell line (bEnd5) as an in vitro blood-brain barrier (BBB) model. The morphology of bEnd5 cells was evaluated using both high-resolution scanning electron microscopy and immunofluorescence to evaluate treatment with depolymerizing agents Cytochalasin D for F-actin filaments and Nocodazole for α-tubulin microtubules. The effects of the depolymerizing agents were investigated on bEnd5 monolayer permeability by measuring the transendothelial electrical resistance (TEER). The data endorsed that during barrier-genesis, F-actin and α-tubulin play a cytoarchitectural role in providing both cell shape dynamics and cytoskeletal structure to TENTs forming across the paracellular space to provide cell-cell engagement. Western blot analysis of the treatments suggested a reduced expression of both proteins, coinciding with a reduction in the rates of cellular proliferation and decreased TEER. The findings endorsed that TENTs provide alignment of the paracellular (PC) spaces and tight junction (TJ) zones to occlude bEnd5 PC spaces. The identification of specific cytoskeletal structures in TENTs endorsed the postulate of their indispensable role in barrier-genesis and the maintenance of regulatory permeability across the BBB.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/ultraestrutura , Proteínas do Citoesqueleto/metabolismo , Actinas/metabolismo , Animais , Biomarcadores , Linhagem Celular , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Imunofluorescência , Expressão Gênica , Camundongos , Nocodazol/farmacologia , Permeabilidade/efeitos dos fármacos
12.
Toxicol Lett ; 354: 56-64, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34757176

RESUMO

The ototoxic mechanisms of cisplatin on the organ of Corti and spiral ganglion neurons have been extensively studied, while few studies have been focused on the stria vascularis (SV). Herein, we verified the functional and morphological impairment in SV induced by a single injection of cisplatin (12 mg/kg, I.P.), represented by a reduction in Endocochlear Potentials (EP) and strial atrophy, and explored underlying mechanisms. Our results revealed increased extravasation of chromatic tracers (Evans blue dye and FITC-dextran) around microvessels after cisplatin exposure. The increased vascular permeability could be attributed to changes of pericytes (PCs) and perivascular-resident macrophage-like melanocytes (PVM/Ms) in number or morphology, as well as the enhanced level of HIF-1α and downstream VEGF. This capillary leakage led to a high accumulation of cisplatin in the perivascular space in SV, and disrupted the integrity of blood-labyrinth barrier (BLB). Also, tight junction (ZO-1) loosening and Na+, K+-ATPase damage was considered to be other critical contributors of BLB breakdown, which resulted in EP drop and consequent hearing loss. This study explored the role of stria vascularis in cisplatin-induced ototoxicity in terms of BLB hyperpermeability and pointed to a novel therapeutic target for the prevention of cisplatin-related hearing loss.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Cóclea/irrigação sanguínea , Cóclea/efeitos dos fármacos , Ototoxicidade/etiologia , Permeabilidade/efeitos dos fármacos , Estria Vascular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Masculino , Camundongos
13.
J Nutr Biochem ; 100: 108886, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34670110

RESUMO

Oxidative stress is directly implicated in the loss of intestinal epithelial barrier function (IEBF) induced by non-steroidal anti-inflammatory drugs (NSAIDs). Previous studies by our research team demonstrated that 2-(3,4-dihydroxybenzoyl)-2,4,6-trihydroxy-3(2H)-benzofuranone (BZF), a quercetin oxidation metabolite that naturally occurs in onion peels, exhibits an antioxidant potency notably higher than quercetin. Thus, we assessed the potential of BZF and a BZF-rich onion peel aqueous extract (OAE) to protect against the loss of IEBF in Caco-2 cell monolayers and in rats exposed to indomethacin. In vitro, pure BZF and OAE standardized in BZF (100 nM), protected against the drop in transepithelial electrical resistance by 70 - 73%. Likewise, it prevented the increase in fluorescein-isothiocyanate labelled dextran (FITC-dextran) paracellular transport by 74% and oxidative stress by 84 - 86%. In vivo, BZF, given orally at a dose 80 µg/Kg bw as OAE, totally abolished a 30-fold increase in FITC-dextran serum concentration induced by indomethacin. This effect was dose-dependent and largely conserved (85%) when OAE was given 180-min prior to indomethacin. The IEBF-protective effect of OAE was accompanied by a full prevention of the NF-ĸB activation, and the increases in interleukine-8 secretion and myeloperoxidase activity induced by indomethacin. The protection was also associated with a 21-fold increase in Nrf2, and a 7-fold and 9-fold increase in heme oxygenase-1 and NAD(P)H-quinone oxidoreductase 1, respectively. The IEBF-protecting effect of OAE involves, most likely, its dual capacity to activate Nrf2 while inhibiting NF-ĸB activation. The extremely low doses of BZF needed to promote such actions warrants extending its IEBF-protective effects to other NSAIDs.


Assuntos
Benzofuranos/farmacologia , Indometacina/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Cebolas/química , Extratos Vegetais/farmacologia , Quercetina/metabolismo , Animais , Anti-Inflamatórios não Esteroides/toxicidade , Células CACO-2 , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/fisiologia , Humanos , Interleucina-8/metabolismo , Mucosa Intestinal/fisiologia , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Oxirredução , Permeabilidade/efeitos dos fármacos , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley
14.
FASEB J ; 36(1): e22061, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34861075

RESUMO

Corticosterone, the stress hormone, exacerbates alcohol-associated tissue injury, but the mechanism involved is unknown. We examined the role of the glucocorticoid receptor (GR) in corticosterone-mediated potentiation of alcohol-induced gut barrier dysfunction and systemic response. Hepatocyte-specific GR-deficient (GRΔHC ) and intestinal epithelial-specific GR-deficient (GRΔIEC ) mice were fed ethanol, combined with corticosterone treatment. Intestinal epithelial tight junction integrity, mucosal barrier function, microbiota dysbiosis, endotoxemia, systemic inflammation, liver damage, and neuroinflammation were assessed. Corticosterone potentiated ethanol-induced epithelial tight junction disruption, mucosal permeability, and inflammatory response in GRΔHC mouse colon; these effects of ethanol and corticosterone were absent in GRΔIEC mice. Gut microbiota compositions in ethanol-fed GRΔHC and GRΔIEC mice were similar to each other. However, corticosterone treatment in ethanol-fed mice shifted the microbiota composition to distinctly different directions in GRΔHC and GRΔIEC mice. Ethanol and corticosterone synergistically elevated the abundance of Enterobacteriaceae and Escherichia coli and reduced the abundance of Lactobacillus in GRΔHC mice but not in GRΔIEC mice. In GRΔHC mice, corticosterone potentiated ethanol-induced endotoxemia and systemic inflammation, but these effects were absent in GRΔIEC mice. Interestingly, ethanol-induced liver damage and its potentiation by corticosterone were observed in GRΔHC mice but not in GRΔIEC mice. GRΔIEC mice were also resistant to ethanol- and corticosterone-induced inflammatory response in the hypothalamus. These data indicate that the intestinal epithelial GR plays a central role in alcohol- and corticosterone-induced gut barrier dysfunction, microbiota dysbiosis, endotoxemia, systemic inflammation, liver damage, and neuroinflammation. This study identifies a novel target for potential therapeutic for alcohol-associated tissue injury.


Assuntos
Corticosterona/efeitos adversos , Etanol/efeitos adversos , Mucosa Intestinal/metabolismo , Receptores de Glucocorticoides/metabolismo , Junções Íntimas/metabolismo , Animais , Corticosterona/farmacologia , Escherichia coli/metabolismo , Etanol/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Lactobacillus/metabolismo , Camundongos , Camundongos Transgênicos , Permeabilidade/efeitos dos fármacos , Receptores de Glucocorticoides/genética , Junções Íntimas/genética
16.
Inflammation ; 45(1): 116-128, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34494203

RESUMO

Angioedema is characterized by swelling of the skin or mucous membranes. Overproduction of the vasodilator bradykinin (BK) is an important contributor to the disease pathology, which causes rapid increase in vascular permeability. BK formation on endothelial cells results from high molecular weight kininogen (HK) interacting with gC1qR, the receptor for the globular heads of C1q, the first component of the classical pathway of complement. Endothelial cells are sensitive to blood-flow-induced shear stress and it has been shown that shear stress can modulate gC1qR expression. This study aimed to determine the following: (1) how BK or angioedema patients' (HAE) plasma affected endothelial cell permeability and gC1qR expression under shear stress, and (2) if monoclonal antibody (mAb) 74.5.2, which recognizes the HK binding site on gC1qR, had an inhibitory effect in HK binding to endothelial cells. Human dermal microvascular endothelial cells (HDMECs) grown on Transwell inserts were exposed to shear stress in the presence of HAE patients' plasma. Endothelial cell permeability was measured using FITC-conjugated bovine serum albumin. gC1qR expression and HK binding to endothelial cell surface was measured using solid-phase ELISA. Cell morphology was quantified using immunofluorescence microscopy. The results demonstrated that BK at 1 µg/mL, but not HAE patients' plasma and/or shear stress, caused significant increases in HDMEC permeability. The mAb 74.5.2 could effectively inhibit HK binding to recombinant gC1qR, and reduce HAE patients' plasma-induced HDMEC permeability change. These results suggested that monoclonal antibody to gC1qR, i.e., 74.5.2, could be potentially used as an effective therapeutic reagent to prevent angioedema.


Assuntos
Angioedema/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Bradicinina/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Proteínas de Transporte/imunologia , Células Endoteliais/efeitos dos fármacos , Proteínas Mitocondriais/imunologia , Angioedema/imunologia , Angioedema/metabolismo , Angioedema/fisiopatologia , Anticorpos Monoclonais/uso terapêutico , Biomarcadores/metabolismo , Permeabilidade Capilar/imunologia , Fármacos Cardiovasculares/uso terapêutico , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Humanos , Permeabilidade/efeitos dos fármacos , Resistência ao Cisalhamento/efeitos dos fármacos
17.
Toxins (Basel) ; 13(12)2021 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-34941701

RESUMO

The fumonisins are a group of common mycotoxins found around the world that mainly contaminate maize. As environmental toxins, they pose a threat to human and animal health. Fumonisin B1 (FB1) is the most widely distributed and the most toxic. FB1 can cause pulmonary edema in pigs. However, the current toxicity mechanism of fumonisins is still in the exploratory stage, which may be related to sphingolipid metabolism. Our study is designed to investigate the effect of FB1 on the cell proliferation and barrier function of swine umbilical vein endothelial cells (SUVECs). We show that FB1 can inhibit the cell viability of SUVECs. FB1 prevents cells from entering the S phase from the G1 phase by regulating the expression of the cell cycle-related genes cyclin B1, cyclin D1, cyclin E1, Cdc25c, and the cyclin-dependent kinase-4 (CDK-4). This results in an inhibition of cell proliferation. In addition, FB1 can also change the cell morphology, increase paracellular permeability, destroy tight junctions and the cytoskeleton, and reduce the expression of tight junction-related genes claudin 1, occludin, and ZO-1. This indicates that FB1 can cause cell barrier dysfunction of SUVECs and promote the weakening or even destruction of the connections between endothelial cells. In turn, this leads to increased blood vessel permeability and promotes exudation. Our findings suggest that FB1 induces toxicity in SUVECs by affecting cell proliferation and disrupting the barrier function.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fumonisinas/toxicidade , Animais , Linhagem Celular , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Suínos , Junções Íntimas/efeitos dos fármacos , Veias Umbilicais/efeitos dos fármacos
18.
Eur J Pharmacol ; 912: 174581, 2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34743979

RESUMO

Sepsis is a systemic inflammatory response syndrome caused by a host's immune response to infection. Acute lung injury (ALI) is one of the most common complications of sepsis with high mortality and morbidity. Recent evidence demonstrated that the 'gut-lung axis' was related to the progression of septic acute lung injury, which regarded gut microbiota and intestinal barrier as two critical factors correlated with acute lung injury. Sinomenine is an isoquinoline alkaloid component extracted from Sinomenium acutum Rehd,et Wils, which has been already reported to have significant anti-inflammatory, immunosuppressive, and anti-arthritis properties. In this research, we observed that sinomenine could repair the lung injury and alleviate inflammatory response induced by cecum ligation and puncture (CLP). Illumine sequencing of 16S rDNA revealed that sinomenine could improve the richness of gut microbiota and modulate the composition of intestinal flora in cecum ligation and puncture mice. Meanwhile, sinomenine could reduce the colon pathological damage and improve the intestine barrier integrity in cecum ligation and puncture mice. We also found that the molecular mechanism of sinomenine's protective effect on intestinal tract was related to the activation of aryl hydrocarbon receptor/nuclear factor erythroid-2 related factor 2(Nrf2)pathway both in vivo and vitro experiments. Collectively, the prevention of septic acute lung injury by sinomenine might be mediated by modulating gut microbiota and restoring intestinal barrier via aryl hydrocarbon receptor/Nrf2-dependent pathway.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Trato Gastrointestinal/metabolismo , Morfinanos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Substâncias Protetoras/farmacologia , Receptores de Hidrocarboneto Arílico/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Animais , Anti-Inflamatórios/uso terapêutico , Ceco/cirurgia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Homeostase/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Ligadura , Masculino , Camundongos Endogâmicos ICR , Morfinanos/uso terapêutico , Permeabilidade/efeitos dos fármacos , Substâncias Protetoras/uso terapêutico , Punções
19.
Front Immunol ; 12: 749002, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759928

RESUMO

Graft-versus-host disease (GVHD) remains a major complication after allogeneic hematopoietic stem cell transplantation (HSCT). An impaired intestinal epithelial barrier is an important component of GVHD pathogenesis. However, contributing host factors that modulate mucosal barrier integrity during GVHD are poorly defined. We hypothesized that vitamin A and retinoic acid (RA) exert positive impacts on maintaining intestinal barrier function after HSCT, thus preventing or dampening GVHD severity. Unexpectedly, we found that exogenous RA increased intestinal permeability of recipient mice after allogeneic HSCT. Serum bacterial endotoxin levels were significantly higher in GVHD mice fed a vitamin A-high (VAH) diet compared to those fed a vitamin A-normal (VAN) diet, indicating a more compromised intestinal barrier function. Furthermore, VAH mice showed more severe lung GVHD with increased donor T cell infiltration in this tissue and died significantly faster than VAN recipients. 16S rRNA sequencing of fecal samples revealed significant differences in the diversity and composition of gut microbiota between VAN and VAH transplant recipients. Collectively, we show that retinoic acid signaling may negatively impact intestinal barrier function during GVHD. Mild vitamin A supplementation is associated with increased lung GVHD and more profound gut dysbiosis. Micronutrients such as vitamin A could modulate complications of allogeneic HSCT, which may be mediated by shaping gut microbiota.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Mucosa Intestinal/efeitos dos fármacos , Vitamina A/farmacologia , Vitaminas/farmacologia , Animais , Células CACO-2 , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Doença Enxerto-Hospedeiro , Humanos , Mucosa Intestinal/metabolismo , Pulmão/efeitos dos fármacos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Permeabilidade/efeitos dos fármacos , RNA Ribossômico 16S , Transdução de Sinais/efeitos dos fármacos , Transplante Homólogo
20.
Angew Chem Int Ed Engl ; 60(52): 26994-27004, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34643312

RESUMO

Lysosome-relevant cell death induced by lysosomal membrane permeabilization (LMP) has recently attracted increasing attention. However, nearly no studies show that currently available LMP inducers can evoke immunogenic cell death (ICD) or convert immunologically cold tumors to hot. Herein, we report a LMP inducer named TPE-Py-pYK(TPP)pY, which can respond to alkaline phosphatase (ALP), leading to formation of nanoassembies along with fluorescence and singlet oxygen turn-on. TPE-Py-pYK(TPP)pY tends to accumulate in ALP-overexpressed cancer cell lysosomes as well as induce LMP and rupture of lysosomal membranes to massively evoke ICD. Such LMP-induced ICD effectively converts immunologically cold tumors to hot as evidenced by abundant CD8+ and CD4+ T cells infiltration into the cold tumors. Exposure of ALP-catalyzed nanoassemblies in cancer cell lysosomes to light further intensifies the processes of LMP, ICD and cold-to-hot tumor conversion. This work thus builds a new bridge between lysosome-relevant cell death and cancer immunotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Morte Celular Imunogênica/efeitos dos fármacos , Lisossomos/metabolismo , Neoplasias/tratamento farmacológico , Organofosfatos/uso terapêutico , Fosfatase Alcalina/metabolismo , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Antineoplásicos/efeitos da radiação , Linhagem Celular Tumoral , Desenho de Fármacos , Células HEK293 , Humanos , Radical Hidroxila/metabolismo , Membranas Intracelulares/metabolismo , Luz , Lisossomos/enzimologia , Camundongos , Organofosfatos/síntese química , Organofosfatos/metabolismo , Organofosfatos/efeitos da radiação , Permeabilidade/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...