Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.215
1.
Sci Rep ; 14(1): 10193, 2024 05 03.
Article En | MEDLINE | ID: mdl-38702361

Amphibians are often recognized as bioindicators of healthy ecosystems. The persistence of amphibian populations in heavily contaminated environments provides an excellent opportunity to investigate rapid vertebrate adaptations to harmful contaminants. Using a combination of culture-based challenge assays and a skin permeability assay, we tested whether the skin-associated microbiota may confer adaptive tolerance to tropical amphibians in regions heavily contaminated with arsenic, thus supporting the adaptive microbiome principle and immune interactions of the amphibian mucus. At lower arsenic concentrations (1 and 5 mM As3+), we found a significantly higher number of bacterial isolates tolerant to arsenic from amphibians sampled at an arsenic contaminated region (TES) than from amphibians sampled at an arsenic free region (JN). Strikingly, none of the bacterial isolates from our arsenic free region tolerated high concentrations of arsenic. In our skin permeability experiment, where we tested whether a subset of arsenic-tolerant bacterial isolates could reduce skin permeability to arsenic, we found that isolates known to tolerate high concentrations of arsenic significantly reduced amphibian skin permeability to this metalloid. This pattern did not hold true for bacterial isolates with low arsenic tolerance. Our results describe a pattern of environmental selection of arsenic-tolerant skin bacteria capable of protecting amphibians from intoxication, which helps explain the persistence of amphibian populations in water bodies heavily contaminated with arsenic.


Amphibians , Arsenic , Microbiota , Skin , Animals , Arsenic/metabolism , Arsenic/toxicity , Microbiota/drug effects , Skin/microbiology , Skin/drug effects , Skin/metabolism , Amphibians/microbiology , Bacteria/drug effects , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Permeability/drug effects
2.
Nutrients ; 16(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732615

Adequate diet, physical activity, and dietary supplementation with muscle-targeted food for special medical purposes (FSMP) or dietary supplement (DS) are currently considered fundamental pillars in sarcopenia treatment. The aim of this study is to evaluate the effectiveness of a DS (containing hydroxy-methyl-butyrate, carnosine, and magnesium, for its action on muscle function and protein synthesis and butyrate and lactoferrin for their contribution to the regulation of gut permeability and antioxidant/anti-inflammation activity) on muscle mass (assessed by dual X-ray absorptiometry (DXA)), muscle function (by handgrip test, chair test, short physical performance battery (SPPB) test, and walking speed test), inflammation (tumor necrosis factor-alpha (TNF-a), C-reactive protein (CRP), and visceral adipose tissue (VAT)) and gut axis (by zonulin). A total of 59 participants (age 79.7 ± 4.8 years, body mass index 20.99 ± 2.12 kg/m2) were enrolled and randomly assigned to intervention (n = 30) or placebo (n = 28). The skeletal muscle index (SMI) significantly improved in the supplemented group compared to the placebo one, +1.02 (CI 95%: -0.77; 1.26), p = 0.001; a significant reduction in VAT was observed in the intervention group, -70.91 g (-13.13; -4.70), p = 0.036. Regarding muscle function, all the tests significantly improved (p = 0.001) in the supplemented group compared to the placebo one. CRP, zonulin, and TNF-alpha significantly decreased (p = 0.001) in intervention, compared to placebo, -0.74 mg/dL (CI 95%: -1.30; -0.18), -0.30 ng/mL (CI 95%: -0.37; -0.23), -6.45 pg/mL (CI 95%: -8.71; -4.18), respectively. This DS improves muscle mass and function, and the gut muscle has emerged as a new intervention target for sarcopenia.


Carnosine , Dietary Supplements , Lactoferrin , Magnesium , Muscle, Skeletal , Permeability , Sarcopenia , Humans , Male , Aged , Female , Sarcopenia/drug therapy , Sarcopenia/prevention & control , Carnosine/administration & dosage , Lactoferrin/administration & dosage , Lactoferrin/pharmacology , Magnesium/administration & dosage , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Permeability/drug effects , Aged, 80 and over , Valerates/administration & dosage , Valerates/pharmacology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Butyrates , Double-Blind Method , Haptoglobins , C-Reactive Protein/metabolism , C-Reactive Protein/analysis , Protein Precursors
3.
Int Immunopharmacol ; 133: 112099, 2024 May 30.
Article En | MEDLINE | ID: mdl-38643709

Visceral hypersensitivity resulting from compromised gut barrier with activated immune system is a key feature of irritable bowel syndrome (IBS). Corticotropin-releasing factor (CRF) and Toll-like receptor 4 (TLR4) activate proinflammatory cytokine signaling to induce these changes, which is one of the mechanisms of IBS. As activation of the NLRP3 inflammasome by lipopolysaccharide (LPS) or TLR4 leads to release interleukin (IL)-1ß, the NLRP3 inflammasome may be involved in the pathophysiology of IBS. Tranilast, an anti-allergic drug has been demonstrated to inhibit the NLRP3 inflammasome, and we evaluated the impact of tranilast on visceral hypersensitivity and colonic hyperpermeability induced by LPS or CRF (IBS rat model). Visceral pain threshold caused by colonic balloon distention was measured by monitoring abdominal muscle contractions electrophysiologically. Colonic permeability was determined by quantifying the absorbed Evans blue within the colonic tissue. Colonic protein levels of NLRP3 and IL-1ß were assessed by immunoblot or ELISA. Intragastric administration of tranilast (20-200 mg/kg) for 3 days inhibited LPS (1 mg/kg)-induced visceral hypersensitivity and colonic hyperpermeability in a dose-dependent manner. Simultaneously, tranilast also abolished these alterations induced by CRF (50 µg/kg). LPS increased colonic protein levels of NLRP3 and IL-1ß, and tranilast inhibited these changes. ß-hydroxy butyrate, an NLRP3 inhibitor, also abolished visceral hypersensitivity and colonic hyperpermeability caused by LPS. In contrast, IL-1ß induced similar GI alterations to LPS, which were not modified by tranilast. In conclusion, tranilast improved visceral pain and colonic barrier by suppression of the NLRP3 inflammasome in IBS rat models. Tranilast may be useful for IBS treating.


Colon , Disease Models, Animal , Inflammasomes , Interleukin-1beta , Irritable Bowel Syndrome , Lipopolysaccharides , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , ortho-Aminobenzoates , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/metabolism , Colon/drug effects , Colon/metabolism , Male , Inflammasomes/metabolism , Inflammasomes/drug effects , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/therapeutic use , Interleukin-1beta/metabolism , Rats , Permeability/drug effects , Hyperalgesia/drug therapy , Visceral Pain/drug therapy , Visceral Pain/metabolism
4.
Int Immunopharmacol ; 133: 112140, 2024 May 30.
Article En | MEDLINE | ID: mdl-38669952

BACKGROUND: Inflammation-induced intestinal barrier dysfunction is not only a pathological feature of Crohn's disease (CD) but also an important therapeutic target. Sclareol (SCL) is a nontoxic natural plant compound with anti-inflammatory effect, but its role in CD has not been established. METHODS: In vivo studies of mice with TNBS-induced colitis were carried out to evaluate the effects of SCL on CD-like colitis and intestinal barrier function. In vitro, a TNF-α-induced colonic organoid model was established to test the direct effect of SCL on inflammation-induced intestinal barrier injure and inflammatory response. The Nrf2/NF-κB/MLCK signalling was analysed to explore the mechanism of SCL. RESULTS: In vivo, SCL largely alleviated the colitis in TNBS mice, as evidenced by improvements in the weight loss, colitis symptoms, endoscopic score, macroscopic histological score, and histological inflammation score. Moreover, SCL significantly improved intestinal barrier dysfunction, manifested as reduced intestinal permeability and decreased intestinal bacterial translocation in TNBS mice. Importantly, SCL antagonised the intestinal mucosal inflammation while protecting tight junctions in TNBS mice. In vitro, SCL largely depressed pro-inflammatory cytokines levels and improved intestinal epithelial permeability in a TNF-α-induced colonic organoid model. In the context of CD, the protective effects of SCL against inflammation and intestinal barrier damage are at least partially results from the Nrf2 signalling activation and the NF-κB/MLCK signalling inhibition. CONCLUSIONS: SCL improved intestinal barrier dysfunction and alleviated CD-like colitis, possibly through modulation of Nrf2/NF-κB/MLCK signalling. In view of SCL's safety profile, there is hope that it will be useful in the clinic.


Colitis , Crohn Disease , Intestinal Mucosa , NF-E2-Related Factor 2 , NF-kappa B , Signal Transduction , Trinitrobenzenesulfonic Acid , Animals , NF-E2-Related Factor 2/metabolism , Crohn Disease/drug therapy , Crohn Disease/pathology , Signal Transduction/drug effects , NF-kappa B/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Humans , Male , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Myosin-Light-Chain Kinase/metabolism , Mice, Inbred C57BL , Permeability/drug effects , Colon/pathology , Colon/drug effects , Diterpenes/therapeutic use , Diterpenes/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
J Ethnopharmacol ; 330: 118194, 2024 Aug 10.
Article En | MEDLINE | ID: mdl-38641077

ETHNOPHARMACOLOGICAL RELEVANCE: Prinsepia utilis Royle, native to the Himalayan region, has a long history of use in traditional medicine for its heat-clearing, detoxification, anti-inflammatory, and analgesic properties. Oils extracted from P. utilis seeds are also used in cooking and cosmetics. With the increasing market demand, this extraction process generates substantial industrial biowastes. Recent studies have found many health benefits with using aqueous extracts of these biowastes, which are also rich in polysaccharides. However, there is limited research related to the reparative effects of the water extracts of P. utilis oil cakes (WEPUOC) on disruptions of the skin barrier function. AIM OF THE STUDY: This study aimed to evaluate the reparative efficacy of WEPUOC in both acute and chronic epidermal permeability barrier disruptions. Furthermore, the study sought to explore the underlying mechanisms involved in repairing the epidermal permeability barrier. MATERIALS AND METHODS: Mouse models with induced epidermal disruptions, employing tape-stripping (TS) and acetone wiping (AC) methods, were used. The subsequent application of WEPUOC (100 mg/mL) was evaluated through various assessments, with a focus on the upregulation of mRNA and protein expression of Corneocyte Envelope (CE) related proteins, lipid synthase-associated proteins, and tight junction proteins. RESULTS: The polysaccharide was the major phytochemicals of WEPUOC and its content was determined as 32.2% by the anthranone-sulfuric acid colorimetric method. WEPUOC significantly reduced transepidermal water loss (TEWL) and improved the damaged epidermal barrier in the model group. Mechanistically, these effects were associated with heightened expression levels of key proteins such as FLG (filaggrin), INV (involucrin), LOR (loricrin), SPT, FASN, HMGCR, Claudins-1, Claudins-5, and ZO-1. CONCLUSIONS: WEPUOC, obtained from the oil cakes of P. utilis, is rich in polysaccharides and exhibits pronounced efficacy in repairing disrupted epidermal barriers through increased expression of critical proteins involved in barrier integrity. Our findings underscore the potential of P. utilis wastes in developing natural cosmetic prototypes for the treatment of diseases characterized by damaged skin barriers, including atopic dermatitis and psoriasis.


Epidermis , Plant Extracts , Tight Junction Proteins , Up-Regulation , Animals , Mice , Plant Extracts/pharmacology , Plant Extracts/chemistry , Tight Junction Proteins/metabolism , Epidermis/drug effects , Epidermis/metabolism , Up-Regulation/drug effects , Water/chemistry , Plant Oils/pharmacology , Plant Oils/chemistry , Male , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/genetics , Permeability/drug effects
6.
Nanotoxicology ; 18(2): 122-133, 2024 Mar.
Article En | MEDLINE | ID: mdl-38436290

Food-grade titanium dioxide (E171) and zinc oxide nanoparticles (ZnO NPs) are found in diverse products for human use. E171 is used as whitening agent in food and cosmetics, and ZnO NPs in food packaging. Their potential multi-organ toxicity has raised concerns on their safety. Since mitochondrial dysfunction is a key aspect of cardio-pathologies, here, we evaluate the effect of chronic exposure to E171 and ZnO NPs in rats on cardiac mitochondria. Changes in cardiac electrophysiology and body weight were measured. E171 reduced body weight more than 10% after 5 weeks. Both E171 and ZnO NPs increased systolic blood pressure (SBP) from 110-120 to 120-140 mmHg after 45 days of treatment. Both NPs altered the mitochondrial permeability transition pore (mPTP), reducing calcium requirement for permeability by 60% and 93% in E171- and ZnO NPs-exposed rats, respectively. Treatments also affected conformational state of adenine nucleotide translocase (ANT). E171 reduced the binding of EMA to Cys 159 in 30% and ZnO NPs in 57%. Mitochondrial aconitase activity was reduced by roughly 50% with both NPs, indicating oxidative stress. Transmission electron microscopy (TEM) revealed changes in mitochondrial morphology including sarcomere discontinuity, edema, and hypertrophy in rats exposed to both NPs. In conclusion, chronic oral exposure to NPs induces functional and morphological damage in cardiac mitochondria, with ZnO NPs being more toxic than E171, possibly due to their dissociation in free Zn2+ ion form. Therefore, chronic intake of these food additives could increase risk of cardiovascular disease.


Mitochondria, Heart , Titanium , Zinc Oxide , Animals , Titanium/toxicity , Zinc Oxide/toxicity , Zinc Oxide/chemistry , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Male , Rats , Administration, Oral , Permeability/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Metal Nanoparticles/toxicity , Nanoparticles/toxicity , Nanoparticles/chemistry , Rats, Sprague-Dawley , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Blood Pressure/drug effects
7.
Prostaglandins Other Lipid Mediat ; 172: 106821, 2024 Jun.
Article En | MEDLINE | ID: mdl-38373554

Endothelial dysfunction often accompanies sepsis. Sevoflurane (Sev) is a widely used inhaled anesthetic that has a protective effect on sepsis-associated damage. We aimed to elucidate the role of Sev in endothelial dysfunction by using a model of LPS induced HUVECs. Sev increased the viability and decreased the apoptosis of HUVECs exposed to LPS. Inflammation and endothelial cell adhesion were improved after Sev addition. Besides, Sev alleviated LPS-induced endothelial cell permeability damage in HUVECs. RORα served as a potential protein that bound to Sev. Importantly, Sev upregulated RORα expression and inhibited endoplasmic reticulum (ER) stress in LPS-treated HUVECs. RORα silencing reversed the impacts of Sev on ER stress. Moreover, RORα deficiency or tunicamycin (ER stress inducer) treatment restored the effects of Sev on the viability, apoptosis, inflammation and endothelial permeability damage of HUVECs exposed to LPS. Taken together, Sev ameliorated LPS-induced endothelial cell damage by targeting RORα to inhibit ER stress.


Apoptosis , Endoplasmic Reticulum Stress , Human Umbilical Vein Endothelial Cells , Inflammation , Lipopolysaccharides , Nuclear Receptor Subfamily 1, Group F, Member 1 , Sevoflurane , Up-Regulation , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Lipopolysaccharides/pharmacology , Endoplasmic Reticulum Stress/drug effects , Apoptosis/drug effects , Sevoflurane/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Inflammation/pathology , Inflammation/metabolism , Inflammation/drug therapy , Up-Regulation/drug effects , Permeability/drug effects
8.
Bioorg Med Chem Lett ; 97: 129486, 2024 01 01.
Article En | MEDLINE | ID: mdl-37734424

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.


Anti-Bacterial Agents , Gram-Negative Bacteria , Anti-Bacterial Agents/chemistry , Drug Discovery , Escherichia coli , Permeability/drug effects , Adenosine/chemistry , Adenosine/pharmacology
9.
Front Immunol ; 14: 1110696, 2023.
Article En | MEDLINE | ID: mdl-36936939

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.


Animal Feed , Bass , Diet , Intestines , Bass/growth & development , Bass/immunology , Bass/physiology , Multiomics , Intestines/chemistry , Intestines/drug effects , Intestines/immunology , Intestines/physiology , Fish Proteins , Animals , Animal Feed/adverse effects , Oxidative Stress/drug effects , Permeability/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Diet/adverse effects , Diet/methods , Diet/veterinary , Fatty Acids/analysis , Cottonseed Oil , Plant Proteins , Chlorella vulgaris , Tenebrio , Edible Insects
10.
Alcohol ; 107: 136-143, 2023 03.
Article En | MEDLINE | ID: mdl-36150609

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.


Dysbiosis , Ethanol , Gastrointestinal Microbiome , Inflammation , Intestines , Animals , Mice , Antimicrobial Peptides/genetics , Antimicrobial Peptides/immunology , Cytokines/immunology , Dysbiosis/chemically induced , Dysbiosis/genetics , Dysbiosis/immunology , Dysbiosis/microbiology , Ethanol/pharmacology , Ethanol/toxicity , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Intestines/drug effects , Intestines/immunology , Intestines/microbiology , Mice, Inbred C57BL , Permeability/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , alpha-Defensins/genetics , alpha-Defensins/immunology
11.
Oxid Med Cell Longev ; 2022: 4834117, 2022.
Article En | MEDLINE | ID: mdl-35251474

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.


Antioxidants/metabolism , Antioxidants/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cysteine/metabolism , Cysteine/pharmacology , Neurodegenerative Diseases/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Biological Transport/drug effects , Excitatory Amino Acid Transporter 3/metabolism , Glutathione/metabolism , Humans , Permeability/drug effects , Prodrugs
12.
Oxid Med Cell Longev ; 2022: 9749461, 2022.
Article En | MEDLINE | ID: mdl-35251482

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.


Cerebral Infarction/drug therapy , Ischemic Stroke/drug therapy , Neuroprotective Agents/therapeutic use , Phytochemicals/therapeutic use , Phytotherapy/methods , Plant Extracts/therapeutic use , Quercetin/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Disease Models, Animal , Humans , Male , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Treatment Outcome
13.
Front Immunol ; 13: 847861, 2022.
Article En | MEDLINE | ID: mdl-35185936

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.


Disulfides/pharmacology , Endoplasmic Reticulum Stress/drug effects , Jejunum/drug effects , Lipopolysaccharides/toxicity , Permeability/drug effects , Sulfinic Acids/pharmacology , Animals , Cell Line , Electric Impedance , Epithelial Cells/metabolism , Heme Oxygenase-1/metabolism , Jejunum/metabolism , Jejunum/physiology , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Swine , Tight Junction Proteins/metabolism
14.
Oxid Med Cell Longev ; 2022: 5044046, 2022.
Article En | MEDLINE | ID: mdl-35222798

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.


Angiotensin II/metabolism , Cardiomegaly/metabolism , Edema, Cardiac/metabolism , Lymphatic Vessels/metabolism , Angiotensin II/administration & dosage , Animals , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Edema, Cardiac/drug therapy , Edema, Cardiac/pathology , Edema, Cardiac/physiopathology , Endothelial Cells/metabolism , Lymphangiogenesis/drug effects , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Permeability/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Vascular Endothelial Growth Factor Receptor-3/deficiency , Vascular Endothelial Growth Factor Receptor-3/metabolism
15.
Mol Med Rep ; 25(4)2022 Apr.
Article En | MEDLINE | ID: mdl-35147204

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.


Donepezil/pharmacology , Myocardial Reperfusion Injury/drug therapy , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Poly (ADP-Ribose) Polymerase-1/metabolism , Reperfusion Injury/drug therapy , Adolescent , Adult , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Endothelial Cells/drug effects , Female , Glucose/metabolism , Humans , Male , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Oxygen/metabolism , Permeability/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Young Adult
16.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article En | MEDLINE | ID: mdl-35054928

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.


Blood-Brain Barrier/metabolism , Blood-Brain Barrier/ultrastructure , Cytoskeletal Proteins/metabolism , Actins/metabolism , Animals , Biomarkers , Cell Line , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Fluorescent Antibody Technique , Gene Expression , Mice , Nocodazole/pharmacology , Permeability/drug effects
17.
Biochim Biophys Acta Biomembr ; 1864(4): 183866, 2022 04 01.
Article En | MEDLINE | ID: mdl-35007517

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.


Colicins/metabolism , Phospholipids/chemistry , Unilamellar Liposomes/metabolism , Amino Acid Sequence , Colicins/chemistry , Colicins/pharmacology , Molecular Dynamics Simulation , Permeability/drug effects , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Protein Conformation, alpha-Helical , Shigella boydii/metabolism , Unilamellar Liposomes/chemistry
18.
Biochem Pharmacol ; 197: 114931, 2022 03.
Article En | MEDLINE | ID: mdl-35085542

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.


KCNQ Potassium Channels/biosynthesis , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Neurons/metabolism , Potassium/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , Glyburide/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neurons/drug effects , Permeability/drug effects , Pregnancy
20.
J Nutr Biochem ; 100: 108886, 2022 02.
Article En | MEDLINE | ID: mdl-34670110

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.


Benzofurans/pharmacology , Indomethacin/toxicity , Intestinal Mucosa/drug effects , Onions/chemistry , Plant Extracts/pharmacology , Quercetin/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Caco-2 Cells , Gastric Mucosa/drug effects , Gastric Mucosa/physiology , Humans , Interleukin-8/metabolism , Intestinal Mucosa/physiology , Male , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidation-Reduction , Permeability/drug effects , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley
...