Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.703
1.
Clin Pharmacol Ther ; 111(1): 252-262, 2022 01.
Article En | MEDLINE | ID: mdl-34564842

Oral alkalization with sodium bicarbonate (NaHCO3 ) or citrate is prescribed for conditions ranging from metabolic acidosis to nephrolithiasis. Although most nephrologists/urologists use this method routinely, extracellular volume (ECV) increase is the main feared adverse event reported for NaHCO3 . Thus far, no trial has specifically studied this issue in a real-world setting. AlcalUN (NCT03035812) is a multicentric, prospective, open-label cohort study with nationwide (France) enrollment in 18 (public and private) nephrology/urology units. Participants were adult outpatients requiring chronic (>1 month) oral alkalization by either NaHCO3 -containing or no-NaHCO3 -containing agents. The ECV increase (primary outcome) was judged based on body weight increase (ΔBW), blood pressure increase (ΔBP), and/or new-onset edema at the first follow-up visit (V1). From February 2017 to February 2020, 156 patients were enrolled. After a median 106 days of treatment, 91 (72%) patients reached the primary outcome. They had lower systolic (135 (125, 141) vs. 141 (130, 150), P = 0.02) and diastolic (77 (67, 85) vs. 85 (73, 90), P = 0.03) BP values, a higher plasma chloride (106.0 (105.0, 109.0) vs. 105.0 (102.0, 107.0), P = 0.02) at baseline, and a less frequent history of nephrolithiasis (32 vs. 56%, P = 0.02). Patients experienced mainly slight ΔBP (< 10 mmHg). The primary outcome was not associated (P = 0.79) with the study treatment (129 received NaHCO3 and 27 received citrate). We subsequently developed three different models of propensity score matching; each confirmed our results. Chronic oral alkalization with NaHCO3 is no longer associated with an ECV increase compared to citrate in real-life settings.


Antacids/adverse effects , Extracellular Space/chemistry , Extracellular Space/drug effects , Sodium Bicarbonate/adverse effects , Aged , Blood Pressure/drug effects , Body Weight/drug effects , Case-Control Studies , Cohort Studies , Female , France , Humans , Male , Middle Aged , Potassium Citrate/adverse effects , Propensity Score , Prospective Studies
2.
Am J Physiol Cell Physiol ; 322(1): C38-C48, 2022 01 01.
Article En | MEDLINE | ID: mdl-34788146

The gaseous signaling molecule hydrogen sulfide (H2S) physiologically regulates store-operated Ca2+ entry (SOCE). The SOCE machinery consists of the plasma membrane-localized Orai channels (Orai1-3) and endoplasmic reticulum-localized stromal interaction molecule (STIM)1 and STIM2 proteins. H2S inhibits Orai3- but not Orai1- or Orai2-mediated SOCE. The current objective was to define the mechanism by which H2S selectively modifies Orai3. We measured SOCE and STIM1/Orai3 dynamics and interactions in HEK293 cells exogenously expressing fluorescently tagged human STIM1 and Orai3 in the presence and absence of the H2S donor GYY4137. Two cysteines (C226 and C232) are present in Orai3 that are absent in the Orai1 and Orai2. When we mutated either of these cysteines to serine, alone or in combination, SOCE inhibition by H2S was abolished. We also established that inhibition was dependent on an interaction with STIM1. To further define the effects of H2S on STIM1/Orai3 interaction, we performed a series of fluorescence recovery after photobleaching (FRAP), colocalization, and fluorescence resonance energy transfer (FRET) experiments. Treatment with H2S did not affect the mobility of Orai3 in the membrane, nor did it influence STIM1/Orai3 puncta formation or STIM1-Orai3 protein-protein interactions. These data support a model in which H2S modification of Orai3 at cysteines 226 and 232 limits SOCE evoked upon store depletion and STIM1 engagement, by a mechanism independent of the interaction between Orai3 and STIM1.


Calcium Channels/metabolism , Calcium Signaling/physiology , Cysteine/metabolism , Hydrogen Sulfide/toxicity , Intracellular Calcium-Sensing Proteins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , HEK293 Cells , Humans , Intracellular Calcium-Sensing Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors
3.
Toxicology ; 458: 152837, 2021 06 30.
Article En | MEDLINE | ID: mdl-34166751

Decabromodiphenyl ether (BDE209) has been widely used as a flame retardant in the past four decades, leading to human health consequences, especially neurological impairments. Our previous in vivo studies have suggested that developmental neurotoxicity in offspring may be the result of BDE209-induced placental type III iodothyronine deiodinase (Dio3) disturbance and consequent thyroid hormone (TH) instability. Dio3 is paternally imprinted gene, and its balanced expression is crucial in directing normal development and growth. In this study, we used placenta-derived cells to investigate how BDE209 affected Dio3 expression through interfering imprinting mechanisms in the delta-like homolog 1 (Dlk1)-Dio3 imprinted region. Gene chip analysis and RT-qPCR identified miR409-3p, miR410-5p, miR494-3p, miR668-3p and miR889-5p as potential candidates involved in Dio3 deregulation. The sodium bisulfite-clonal sequencing revealed the BDE209 affect methylation status of two differentially methylated regions (DMRs), intergenic-DMR (IG-DMR) and maternally expressed gene 3-DMR (MEG3-DMR). Our data indicate that placental Dio3 may be a potential molecular target for future study of BDE209 developmental toxicity. In particular, miRNAs, IG-DMR and MEG3-DMR in the Dlk1-Dio3 imprinted locus may be informative in directing studies in TH disturbance and developmental toxicity induced by in utero exposure to environmental persistent organic pollutants (POPs), and those candidate miRNAs may prove to be convenient and noninvasive biomarkers for future large-scale population studies.


Calcium-Binding Proteins/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Flame Retardants/toxicity , Halogenated Diphenyl Ethers/toxicity , Iodide Peroxidase/drug effects , Membrane Proteins/drug effects , Placenta/drug effects , Placenta/metabolism , Thyroid Hormones/metabolism , Cell Line, Tumor , DNA Methylation , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Developmental , Humans , MicroRNAs/metabolism , Pregnancy , Transfection
4.
ACS Appl Mater Interfaces ; 13(16): 18604-18618, 2021 Apr 28.
Article En | MEDLINE | ID: mdl-33856200

Microfluctuations in a pH gradient create a harsh microenvironment in tumors, leaving behind the most aggressive, invasive, and drug-resistant tumor cells. Directly visualizing the spatiotemporal distribution of pH variations and accurately quantifying the dynamic acid-base changes during cancer treatment are critical to estimate prognosis and to evaluate therapeutic efficacy. However, the quantification of subtle pH variations dynamically and noninvasively remains challenging. The purpose of this study is to determine and visualize dynamic acid-base changes in solid tumors during anti-acid treatments by magnetic resonance imaging (MRI) using pH-sensitive nanoparticles. We report the development of pH-sensitive nanoparticles, MnO2@BSA, that rapidly and strongly amplify the MR contrast signal in response to the extracellular acidic environment of solid tumors. The spatiotemporal distribution and dynamic fluctuations of pH heterogeneity in NCI-H460 lung tumors were observed with MnO2@BSA at different time points after an anti-acid treatment with esomeprazole, which directly interferes with the acidic microenvironment of the tumor. Imaging results were validated using a pH microsensor. MRI of pH-sensitive MnO2@BSA nanoparticles provided direct readouts of the kinetics of pH gradient fluctuations during esomeprazole treatment. A significant MR signal reduction was observed at the 48 h time point after treatment. The manipulated extracellular pH changes detected noninvasively by MRI coincided with the extracellular pH fluctuations measured with a pH microsensor (pH 6.12-6.63). Immunofluorescence and Western blot analyses confirmed the expression of V-ATPase in NCI-H460 lung cancer cells, which could be inhibited by esomeprazole, as detected by ELISA assay. Overall, these results demonstrate that MnO2@BSA MRI has great potential as a noninvasive tool to accurately monitor pH fluctuations, thereby paving the way for the dynamic detection of acidic microenvironments in vivo without the need for pH microsensors.


Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Magnetic Resonance Imaging , Manganese Compounds/chemistry , Nanoparticles/chemistry , Oxides/chemistry , Serum Albumin, Bovine/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Extracellular Space/drug effects , Extracellular Space/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Tumor Microenvironment/drug effects
5.
Biochem Biophys Res Commun ; 546: 138-144, 2021 03 26.
Article En | MEDLINE | ID: mdl-33582556

Hepatic injury is common in patients who suffer from severe burns plus delayed resuscitation (B + DR). Stimulator of interferon genes (STING) is primarily expressed in Kupffer cells (KCs). We demonstrated that B + DR caused hepatic injury and oxidative stress. Reactive oxygen species (ROS) damage mitochondrial membranes in hepatocytes, leading to the release of mitochondrial DNA (mtDNA) into the hepatocyte cytosol and the circulation. The damaged hepatocytes then activate the mtDNA/STING pathway in KCs and trigger KCs polarization towards pro-inflammatory phenotype. SS-31 is a strong antioxidant that specifically concentrates in the inner mitochondrial membrane. SS-31 prevented hepatic injury by neutralizing ROS, inhibiting the release of mtDNA, protecting hepatocyte mitochondria, suppressing the activation of the mtDNA/STING pathway and inhibiting KCs polarization into pro-inflammatory phenotype.


Adaptor Proteins, Signal Transducing/metabolism , Burns/complications , DNA, Mitochondrial/drug effects , Kupffer Cells/drug effects , Liver/drug effects , Liver/injuries , Membrane Proteins/metabolism , Oligopeptides/pharmacology , Resuscitation , Animals , DNA, Mitochondrial/blood , DNA, Mitochondrial/metabolism , Extracellular Space/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Kupffer Cells/metabolism , Male , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Time Factors
6.
Biol Pharm Bull ; 44(2): 266-270, 2021.
Article En | MEDLINE | ID: mdl-33518679

Extracellular pH (pHe) of tumor cells is characteristic of tumor microenvironment (TME). Acidic TME impairs the responses of tumors to some anti-cancer chemotherapies. In this study, we showed that daily oral dosing of sodium potassium citrate (K/Na citrate) increased blood HCO3- concentrations, corresponding to increase of HCO3- concentrations and pHs in urine, and neutralized the tumor pHe. Neutralization of acidic TME by alkaline substance like HCO3-, an active metabolite of K/Na citrate, well potentiated the therapeutic effect of anticancer agent TS-1®, an orally active 5-fuluoro-uracil derivative, in Panc-1 pancreatic cancer-xenograft murine model. Neutralization of acidic TME by using an alkaline K/Na citrate is a smart approach for enhancement of the therapeutic effects of anticancer agents for pancreatic cancer in the end stage.


Antacids/administration & dosage , Hydrogen-Ion Concentration/drug effects , Oxonic Acid/administration & dosage , Pancreatic Neoplasms/drug therapy , Tegafur/administration & dosage , Tumor Microenvironment/drug effects , Administration, Oral , Animals , Antacids/pharmacokinetics , Cell Line, Tumor , Drug Combinations , Drug Synergism , Extracellular Space/chemistry , Extracellular Space/drug effects , Female , Humans , Mice , Oxonic Acid/pharmacokinetics , Oxonic Acid/therapeutic use , Pancreatic Neoplasms/pathology , Potassium Citrate/administration & dosage , Potassium Citrate/pharmacokinetics , Sodium Citrate/administration & dosage , Sodium Citrate/pharmacokinetics , Tegafur/pharmacokinetics , Tegafur/therapeutic use , Xenograft Model Antitumor Assays
7.
Int J Med Sci ; 18(3): 652-659, 2021.
Article En | MEDLINE | ID: mdl-33437200

Objective: Different anesthetics have distinct effects on the interstitial fluid (ISF) drainage in the extracellular space (ECS) of the superficial rat brain, while their effects on ISF drainage in the ECS of the deep rat brain still remain unknown. Herein, we attempt to investigate and compare the effects of propofol and isoflurane on ECS structure and ISF drainage in the caudate-putamen (CPu) and thalamus (Tha) of the deep rat brain. Methods: Adult Sprague-Dawley rats were anesthetized with propofol or isoflurane, respectively. Twenty-four anesthetized rats were randomly divided into the propofol-CPu, isoflurane-CPu, propofol-Tha, and isoflurane-Tha groups. Tracer-based magnetic resonance imaging (MRI) and fluorescent-labeled tracer assay were utilized to quantify ISF drainage in the deep brain. Results: The half-life of ISF in the propofol-CPu and propofol-Tha groups was shorter than that in the isoflurane-CPu and isoflurane-Tha groups, respectively. The ECS volume fraction in the propofol-CPu and propofol-Tha groups was much higher than that in the isoflurane-CPu and isoflurane-Tha groups, respectively. However, the ECS tortuosity in the propofol-CPu and propofol-Tha groups was much smaller than that in isoflurane-CPu and isoflurane-Tha groups, respectively. Conclusions: Our results demonstrate that propofol rather than isoflurane accelerates the ISF drainage in the deep rat brain, which provides novel insights into the selective control of ISF drainage and guides selection of anesthetic agents in different clinical settings, and unravels the mechanism of how general anesthetics function.


Anesthetics, General/administration & dosage , Caudate Nucleus/drug effects , Extracellular Fluid/metabolism , Putamen/drug effects , Thalamus/drug effects , Administration, Inhalation , Animals , Caudate Nucleus/cytology , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Gadolinium DTPA/administration & dosage , Infusions, Parenteral , Isoflurane/administration & dosage , Magnetic Resonance Imaging/methods , Models, Animal , Propofol/administration & dosage , Putamen/cytology , Putamen/diagnostic imaging , Putamen/metabolism , Rats , Rats, Sprague-Dawley , Thalamus/cytology , Thalamus/diagnostic imaging , Thalamus/metabolism
8.
ACS Appl Mater Interfaces ; 12(52): 57798-57809, 2020 Dec 30.
Article En | MEDLINE | ID: mdl-33325679

Metastasis is the primary cause of death for most cancer patients, in which tumor-associated macrophages (TAMs) are involved through several mechanisms. While hitherto there is still a lack of study on exclusive elimination of TAMs to inhibit metastasis due to the difficulties in specific targeting of TAMs, we construct an extra- and intracellular stepwise-responsive delivery system p-(aminomethyl)benzoic acid (PAMB)/doxorubicin (DOX) to achieve specific TAM depletion for the first time, thereby preventing tumor metastasis. Once accumulated into the tumor, PAMB/DOX would stepwise responsively (hypoxia and reactive oxygen species (ROS) responsively) disintegrate to expose the TAM-targeting ligand and release DOX sequentially, which depletes TAMs effectively in vivo. Owing to the inhibition of extracellular matrix (ECM) degradation, neovascularization, and tumor invasion contributed by TAM depletion, lung metastasis was successfully inhibited. Furthermore, PAMB/DOX showed efficient inhibition against tumor growth as well as spontaneous metastasis formation when combined with additional chemotherapy, representing a safe and efficient nanoplatform to modulate the adverse tumor microenvironment via TAM elimination.


Drug Carriers/chemistry , Extracellular Space/drug effects , Intracellular Space/drug effects , Nanostructures/chemistry , Tumor-Associated Macrophages/drug effects , para-Aminobenzoates/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Liberation , Extracellular Space/metabolism , Intracellular Space/metabolism , Mice , Neoplasm Metastasis , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Tissue Distribution , Tumor Hypoxia/drug effects , Tumor-Associated Macrophages/pathology , para-Aminobenzoates/pharmacokinetics
9.
J Neurosci ; 40(49): 9440-9454, 2020 12 02.
Article En | MEDLINE | ID: mdl-33122389

Understanding the effects of anesthesia on cortical neuronal spiking and information transfer could help illuminate the neuronal basis of the conscious state. Recent investigations suggest that the brain state identified by local field potential spectrum is not stationary but changes spontaneously at a fixed level of anesthetic concentration. How cortical unit activity changes with dynamically transitioning brain states under anesthesia is unclear. Extracellular unit activity was measured with 64-channel silicon microelectrode arrays in cortical layers 5/6 of the primary visual cortex of chronically instrumented, freely moving male rats (n = 7) during stepwise reduction of the anesthetic desflurane (6%, 4%, 2%, and 0%). Unsupervised machine learning applied to multiunit spike patterns revealed five distinct brain states. A novel desynchronized brain state with increased spike rate variability, sample entropy, and EMG activity occurred in 6% desflurane with 40.0% frequency. The other four brain states reflected graded levels of anesthesia. As anesthesia deepened the spike rate of neurons decreased regardless of their spike rate profile at baseline conscious state. Actively firing neurons with wide-spiking pattern showed increased bursting activity along with increased spike timing variability, unit-to-population correlation, and unit-to-unit transfer entropy, despite the overall decrease in transfer entropy. The narrow-spiking neurons showed similar changes but to a lesser degree. These results suggest that (1) anesthetic effect on spike rate is distinct from sleep, (2) synchronously fragmented spiking pattern is a signature of anesthetic-induced unconsciousness, and (3) the paradoxical, desynchronized brain state in deep anesthesia contends the generally presumed monotonic, dose-dependent anesthetic effect on the brain.SIGNIFICANCE STATEMENT Recent studies suggest that spontaneous changes in brain state occur under anesthesia. However, the spiking behavior of cortical neurons associated with such state changes has not been investigated. We found that local brain states defined by multiunit activity had a nonunitary relationship with the current anesthetic level. A paradoxical brain state displaying asynchronous firing pattern and high EMG activity was found unexpectedly in deep anesthesia. In contrast, the synchronous fragmentation of neuronal spiking appeared to be a robust signature of the state of anesthesia. The findings challenge the assumption of monotonic, anesthetic dose-dependent behavior of cortical neuron populations. They enhance the interpretation of neuroscientific data obtained under anesthesia and the understanding of the neuronal basis of anesthetic-induced state of unconsciousness.


Anesthesia , Brain/drug effects , Cerebral Cortex/drug effects , Anesthetics, Inhalation/pharmacology , Animals , Desflurane/pharmacology , Dose-Response Relationship, Drug , Electrodes, Implanted , Electroencephalography , Electromyography , Electrophysiological Phenomena/drug effects , Entropy , Extracellular Space/drug effects , Male , Neurons/drug effects , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Sleep/physiology , Unconsciousness/chemically induced , Unconsciousness/physiopathology
10.
ASN Neuro ; 12: 1759091420967152, 2020.
Article En | MEDLINE | ID: mdl-33092407

Astrocytes and neurons have been shown to swell across a variety of different conditions, including increases in extracellular potassium concentration (^[K+]o). The mechanisms involved in the coupling of K+ influx to water movement into cells leading to cell swelling are not well understood and remain controversial. Here, we set out to determine the effects of ^[K+]o on rapid volume responses of hippocampal CA1 pyramidal neurons and stratum radiatum astrocytes using real-time confocal volume imaging. First, we found that elevating [K+]o within a physiological range (to 6.5 mM and 10.5 mM from a baseline of 2.5 mM), and even up to pathological levels (26 mM), produced dose-dependent increases in astrocyte volume, with absolutely no effect on neuronal volume. In the absence of compensating for addition of KCl by removal of an equal amount of NaCl, neurons actually shrank in ^[K+]o, while astrocytes continued to exhibit rapid volume increases. Astrocyte swelling in ^[K+]o was not dependent on neuronal firing, aquaporin 4, the inwardly rectifying potassium channel Kir 4.1, the sodium bicarbonate cotransporter NBCe1, , or the electroneutral cotransporter, sodium-potassium-chloride cotransporter type 1 (NKCC1), but was significantly attenuated in 1 mM barium chloride (BaCl2) and by the Na+/K+ ATPase inhibitor ouabain. Effects of 1 mM BaCl2 and ouabain applied together were not additive and, together with reports that BaCl2 can inhibit the NKA at high concentrations, suggests a prominent role for the astrocyte NKA in rapid astrocyte volume increases occurring in ^[K+]o. These findings carry important implications for understanding mechanisms of cellular edema, regulation of the brain extracellular space, and brain tissue excitability.


Aquaporin 4/metabolism , Astrocytes/metabolism , Cell Size , Hippocampus/metabolism , Potassium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Astrocytes/drug effects , Cell Size/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Potassium/pharmacology
11.
Biol Pharm Bull ; 43(9): 1306-1314, 2020.
Article En | MEDLINE | ID: mdl-32879204

The extracellular space (ECS) is the space between the neurons and the capillaries in the brain. The volume fraction (α) and the tortuosity (λ) are the main parameters used to describe its characteristics. Thymoquinone has been proved to possess anti-oxidant and anti-inflammatory activity. In this study, we used a gadolinium-diethylenetriaminepentacetate (Gd-DTPA)-enhanced magnetic resonance imaging (MRI) system to determine the effects of thymoquinone on ECS parameters in transient middle cerebral artery occlusion rats (tMCAO) to prove the neuroprotective effect of thymoquinone on brain tissue damage caused by ischemic stroke. Neurological examinations, 2,3,5-triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (H&E) staining and assaying of ECS parameters using MRI were performed 24 h after surgery. We found that thymoquinone could improve the behavioural performance by neurological examinations. TTC staining indicated that thymoquinone significantly decreased the percentage of hemi-cerebral infarction. Also, H&E staining showed that thymoquinone could inhibit the neuron necrosis in the hippocampal CA1 region. We found that thymoquinone treatment could inhibit the changes in ECS diffusion parameters, which might prove that thymoquinone might protect brain tissue damage caused by ischemic stroke. Thymoquinone can protect the brain against cerebral ischemia-reperfusion injury, effectively ameliorate abnormalities in characteristics of ECS and decrease cerebral infarction in tMCAO rats.


Benzoquinones/administration & dosage , Brain/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Stroke/drug therapy , Neuroprotective Agents/administration & dosage , Animals , Brain/cytology , Brain/diagnostic imaging , Brain/pathology , Disease Models, Animal , Extracellular Space/diagnostic imaging , Extracellular Space/drug effects , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/diagnosis , Infarction, Middle Cerebral Artery/pathology , Ischemic Stroke/diagnosis , Ischemic Stroke/etiology , Ischemic Stroke/pathology , Magnetic Resonance Imaging , Male , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley
12.
PLoS One ; 15(9): e0238178, 2020.
Article En | MEDLINE | ID: mdl-32946441

Mycobacterium abscessus (M. abscessus), a rapidly growing mycobacterium, is an emergent opportunistic pathogen responsible for chronic bronchopulmonary infections in individuals with respiratory diseases such as cystic fibrosis. Most treatments of M. abscessus pulmonary infections are poorly effective due to the intrinsic resistance of this bacteria against a broad range of antibiotics including anti-tuberculosis agents. Consequently, the number of drugs that are efficient against M. abscessus remains limited. In this context, 19 oxadiazolone (OX) derivatives have been investigated for their antibacterial activity against both the rough (R) and smooth (S) variants of M. abscessus. Several OXs impair extracellular M. abscessus growth with moderated minimal inhibitory concentrations (MIC), or act intracellularly by inhibiting M. abscessus growth inside infected macrophages with MIC values similar to those of imipenem. Such promising results prompted us to identify the potential target enzymes of the sole extra and intracellular inhibitor of M. abscessus growth, i.e., compound iBpPPOX, via activity-based protein profiling combined with mass spectrometry. This approach led to the identification of 21 potential protein candidates being mostly involved in M. abscessus lipid metabolism and/or in cell wall biosynthesis. Among them, the Ag85C protein has been confirmed as a vulnerable target of iBpPPOX. This study clearly emphasizes the potential of the OX derivatives to inhibit the extracellular and/or intracellular growth of M. abscessus by targeting various enzymes potentially involved in many physiological processes of this most drug-resistant mycobacterial species.


Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Mycobacterium abscessus/drug effects , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Animals , Extracellular Space/drug effects , Extracellular Space/microbiology , Intracellular Space/drug effects , Intracellular Space/microbiology , Mice , Microbial Sensitivity Tests , Mycobacterium abscessus/growth & development , RAW 264.7 Cells
13.
Molecules ; 25(16)2020 Aug 06.
Article En | MEDLINE | ID: mdl-32781695

Previous studies have reported that estrogen hormone promotes melanogenesis while progesterone inhibits it. A selective estrogen receptor modulator (SERM), tamoxifen, has been shown to promote melanogenesis; however, to date, there have been no reports on the effects of a selective progesterone receptor modulator (SPRM) on melanogenesis. In the present study, we hypothesized that asoprisnil (AP), a SPRM, inhibits melanogenesis. AP was tested for cytotoxicity to B16F10 mouse melanoma cells for screening the nontoxic concentrations using MTS cytotoxicity assay. Extracellular and intracellular melanin levels were estimated at nontoxic concentrations of AP. To evaluate the direct effect of AP on tyrosinase enzyme, tyrosinase activity and copper chelating activities were measured. Next, the effects of AP on melanogenesis were tested in normal human melanocytes, neonatal, darkly pigmented (HEMn-DP). Our results demonstrate that AP was nontoxic at a concentration range of 10-50 µM in B16F10 cells; AP at 50 µM significantly suppressed extracellular melanin levels comparable to kojic acid at 500 µM, with no significant effect on intracellular melanin levels. The mechanism of melanogenesis inhibition was studied to assess if AP downregulated tyrosinase activity in cell lysates or in a cell-free system. However, AP was found to increase intracellular tyrosinase activity without any effect on tyrosinase enzyme activity or copper chelating activity in a cell-free system, indicating that AP inhibits melanogenesis by mechanisms other than direct effects on tyrosinase enzyme activity. The capacity of AP to inhibit melanosome export was further validated in HEMn-DP cells; AP significantly suppressed dendricity at concentrations of 20 and 30 µM in the absence of effects on melanin synthesis or intracellular tyrosinase activity. In addition, AP was nontoxic to human keratinocytes (HaCaT) at these concentrations, validating its safety for topical use. Taken together, our preliminary results demonstrate that AP might be repurposed as a candidate therapeutic for treatment of hyperpigmentation disorders via a unique mechanism, which encompasses a selective inhibition of melanosome export.


Estrenes/pharmacology , Melanosomes/drug effects , Melanosomes/metabolism , Oximes/pharmacology , Receptors, Progesterone/metabolism , Animals , Biological Transport/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Extracellular Space/drug effects , Extracellular Space/metabolism , Melanoma, Experimental/pathology , Mice
14.
Nucleosides Nucleotides Nucleic Acids ; 39(10-12): 1400-1409, 2020.
Article En | MEDLINE | ID: mdl-32571143

Trehalose is a stable, non-reducing disaccharide, which was found recently to stimulate autophagy, limit the inflammatory response and suppress the growth of specific types of cancer. Purinergic signaling and dysregulation of nucleotide metabolism are the key factors, which play a role in the pathophysiology of cancer development and inflammation. Therefore, this study took a novel approach and aimed to find the effect of trehalose on intracellular, and the extracellular metabolism of nucleotides and NAD + in endothelial and breast cancer cells. The results of this study indicated that in vitro concentrations of trehalose between 0.5 and 5 mM reduced the levels of intracellular NAD + in breast cancer cells. The decrease of intracellular guanosine, independent of GTP energy metabolism, was also observed in both endothelial and cancer cells. Trehalose decreased the activity of ecto-adenosine deaminase. Maximal 3-fold decrease in adenosine deamination was observed in both cell types. Trehalose causes changes in both intracellular and extracellular nucleotide metabolism that is more significant in cancer cells than in endothelium. This effect may have therapeutic potential in cancer and endothelial dysfunction, but its full clarification requires further studies.


Extracellular Space/drug effects , Extracellular Space/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Nucleotides/metabolism , Trehalose/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Pilot Projects
15.
Hipertens. riesgo vasc ; 37(2): 72-77, abr.-jun. 2020. tab
Article Es | IBECS | ID: ibc-189194

El sistema renina-angiotensina (SRA) es una cascada hormonal que regula presión arterial, electrólitos y balance hídrico. La angiotensinaII (AII) ejerce sus efectos a través de los receptores AT1 y AT2. El AT1 se encuentra en el sincitiotrofoblasto; el AT2 predomina durante el desarrollo fetal y su estimulación inhibe el crecimiento celular, aumenta la apoptosis, causa vasodilatación y regula el desarrollo del tejido fetal. Existe además un SRA en la placenta, y la generación local de AII es responsable de la activación de los receptores AT1 del trofoblasto. En el embarazo normal, concomitantemente con reducción de los niveles de presión arterial, se produce un aumento del SRA circulante, pero la presión arterial no sube porque existe refractariedad a la AII, cosa que no ocurre en la preeclampsia. Revisamos la función del SRA en el embarazo normal y en la preeclampsia


The renin-angiotensin system (ARS) is a hormonal cascade that regulates blood pressure, electrolytes and water balance. AngiotensinII (AII) exerts its effects through the AT1 and AT2 receptors. AT1 is found in the syncytiotrophoblast, AT2 predominates during foetal development and its stimulation inhibits cell growth, increases apoptosis, causes vasodilation and regulates the development of foetal tissue. There is also an SRA in the placenta. The local generation of AII is responsible for the activation of AT1 receptors in the trophoblast. In normal pregnancy, concomitantly with reduction of blood pressure the circulating RAS increases, but blood pressure does not rise due to AII refractoriness, which does not occur in preeclampsia. We review the role of the SRA in normal pregnancy and preeclampsia


Humans , Female , Pregnancy , Renin-Angiotensin System/drug effects , Pre-Eclampsia/metabolism , Angiotensin-Converting Enzyme Inhibitors/metabolism , Hemodynamics/drug effects , Serum Albumin/drug effects , Severity of Illness Index , Extracellular Space/drug effects , Homeostasis/drug effects
16.
J Neurosci ; 40(27): 5161-5176, 2020 07 01.
Article En | MEDLINE | ID: mdl-32444385

Alterations of excitatory synaptic function are the strongest correlate to the pathologic disturbance of cognitive ability observed in the early stages of Alzheimer's disease (AD). This pathologic feature is driven by amyloid-ß oligomers (Aßos) and propagates from neuron to neuron. Here, we investigated the mechanism by which Aßos affect the function of synapses and how these alterations propagate to surrounding healthy neurons. We used complementary techniques ranging from electrophysiological recordings and molecular biology to confocal microscopy in primary cortical cultures, and from acute hippocampal and cortical slices from male wild-type and amyloid precursor protein (APP) knock-out (KO) mice to assess the effects of Aßos on glutamatergic transmission, synaptic plasticity, and dendritic spine structure. We showed that extracellular application of Aßos reduced glutamatergic synaptic transmission and long-term potentiation. These alterations were not observed in APP KO neurons, suggesting that APP expression is required. We demonstrated that Aßos/APP interaction increases the amyloidogenic processing of APP leading to intracellular accumulation of newly produced Aßos. Intracellular Aßos participate in synaptic dysfunctions as shown by pharmacological inhibition of APP processing or by intraneuronal infusion of an antibody raised against Aßos. Furthermore, we provide evidence that following APP processing, extracellular release of Aßos mediates the propagation of the synaptic pathology characterized by a decreased spine density of neighboring healthy neurons in an APP-dependent manner. Together, our data unveil a complementary role for Aßos in AD, while intracellular Aßos alter synaptic function, extracellular Aßos promote a vicious cycle that propagates synaptic pathology from diseased to healthy neurons.SIGNIFICANCE STATEMENT Here we provide the proof that a vicious cycle between extracellular and intracellular pools of Aß oligomers (Aßos) is required for the spreading of Alzheimer's disease (AD) pathology. We showed that extracellular Aßos propagate excitatory synaptic alterations by promoting amyloid precursor protein (APP) processing. Our results also suggest that subsequent to APP cleavage two pools of Aßos are produced. One pool accumulates inside the cytosol, inducing the loss of synaptic plasticity potential. The other pool is released into the extracellular space and contributes to the propagation of the pathology from diseased to healthy neurons. Pharmacological strategies targeting the proteolytic cleavage of APP disrupt the relationship between extracellular and intracellular Aß, providing a therapeutic approach for the disease.


Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/metabolism , Neuronal Plasticity/drug effects , Neurons/metabolism , Synapses/drug effects , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Antibodies, Blocking/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Histidine/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Patch-Clamp Techniques , Primary Cell Culture , Synaptic Transmission/drug effects
17.
Neuropharmacology ; 171: 108083, 2020 07.
Article En | MEDLINE | ID: mdl-32304702

Isatin is an endogenous indole that inhibits monoamine oxidase (MAO). When exogenously administered, it increases the striatal dopamine and acetylcholine levels and presents neuroprotective effects in the brain. Previous studies show that intrastriatal administration of isatin increased the in vivo dopamine release from striatum in a concentration-dependent form. In the present work, we investigated the effects of combined administration of isatin together with other substances actually used in antiparkinsonian pharmacotherapy on in vivo dopamine overflow. For this, we co-administered isatin with the MAO inhibitors selegiline and clorgyline, l-DOPA, the catechol-o-methyl-transferase (COMT) inhibitors tropolone and dinitrocatechol, with the dopaminergic agonist ropinirole, and with the psychostimulant caffeine, in order to evaluate possible synergies between these substances to increase the dopamine extracellular levels in freely moving rats. Intrastriatal administration of isatin (10 mM, 60 min) significantly increased dopamine release to 1164 ± 152%, compared to the baseline. Co-administration of isatin together with selegiline (1 mM) or clorgyline (1 mM) alone or in combinations showed a similar profile to increase in vivo dopamine release. Intrastriatal infusion of isatin together with antiparkinsonian drugs l-DOPA (25 µM), tropolone (1 mM), dinitrocatechol (100 µM), amantadine (5 mM) and caffeine (5 mM) significantly elevated extracellular dopamine levels more than any single drug, showing a good neurochemical synergy by improving the effect of isatin on the extracellular dopamine levels in the striatum. Infusion of isatin + ropinirole (5 mM) did not change the isatin-induced increase in dopamine overflow. These results could be useful to carry out further investigations with a possible clinical application.


Antiparkinson Agents/pharmacology , Corpus Striatum/metabolism , Dopamine/metabolism , Isatin/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/physiology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Catechol O-Methyltransferase/drug effects , Central Nervous System Stimulants/pharmacology , Corpus Striatum/drug effects , Dopamine Agonists/pharmacology , Drug Synergism , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Homovanillic Acid/metabolism , Male , Microinjections , Rats , Rats, Sprague-Dawley
18.
Bioelectrochemistry ; 134: 107519, 2020 Aug.
Article En | MEDLINE | ID: mdl-32251985

Extracellular electron transfer (EET) allows microorganisms to perform anaerobic respiration using insoluble electron acceptors, including minerals and electrodes. EET-based applications require efficient electron transfer between living and non-living systems. To improve EET efficiency, the TiO2@TiN nanocomposite was used to form hybrid biofilms with Shewanella loihica PV-4 (PV-4). Chronoamperometry showed that peak current was increased 4.6-fold via the addition of the TiO2@TiN nanocomposite. Different biofilms were further tested in a dual-chamber microbial fuel cell. The PV-4 biofilm resulted a maximum power density of 33.4 mW/m2, while the hybrid biofilm of the TiO2@TiN nanocomposite with PV-4 yielded a 92.8% increase of power density. Electrochemical impedance spectroscopy analyses showed a lower electron-transfer resistance in the hybrid biofilm. Biological measurements revealed that both flavin secretion and cytochrome c expression were increased when the TiO2@TiN nanocomposite presented. These results demonstrated that the TiO2@TiN nanocomposite could synergistically enhance the EET of PV-4 through altering its metabolism. Our findings provide a new strategy for optimizing biotic-abiotic interactions in bioelectrochemical systems.


Extracellular Space/drug effects , Extracellular Space/metabolism , Nanocomposites , Shewanella/cytology , Shewanella/drug effects , Titanium/pharmacology , Biofilms/drug effects , Drug Synergism , Electron Transport/drug effects , Shewanella/physiology
19.
Adv Mater ; 32(19): e1907267, 2020 May.
Article En | MEDLINE | ID: mdl-32182391

Ion homeostasis is essential for cellular survival, and elevated concentrations of specific ions are used to start distinct forms of programmed cell death. However, investigating the influence of certain ions on cells in a controlled way has been hampered due to the tight regulation of ion import by cells. Here, it is shown that lipid-coated iron-based metal-organic framework nanoparticles are able to deliver and release high amounts of iron ions into cells. While high concentrations of iron often trigger ferroptosis, here, the released iron induces pyroptosis, a form of cell death involving the immune system. The iron release occurs only in slightly acidic extracellular environments restricting cell death to cells in acidic microenvironments and allowing for external control. The release mechanism is based on endocytosis facilitated by the lipid-coating followed by degradation of the nanoparticle in the lysosome via cysteine-mediated reduction, which is enhanced in slightly acidic extracellular environment. Thus, a new functionality of hybrid nanoparticles is demonstrated, which uses their nanoarchitecture to facilitate controlled ion delivery into cells. Based on the selectivity for acidic microenvironments, the described nanoparticles may also be used for immunotherapy: the nanoparticles may directly affect the primary tumor and the induced pyroptosis activates the immune system.


Extracellular Space/chemistry , Extracellular Space/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Nanoparticles/chemistry , Pyroptosis/drug effects , HeLa Cells , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Conformation
20.
Sci China Life Sci ; 63(9): 1363-1379, 2020 Sep.
Article En | MEDLINE | ID: mdl-32133594

Brain interstitial fluid drainage and extracellular space are closely related to waste clearance from the brain. Different anesthetics may cause different changes of brain interstitial fluid drainage and extracellular space but these still remain unknown. Herein, effects of the inhalational isoflurane, intravenous sedative dexmedetomidine and pentobarbital sodium on deep brain matters' interstitial fluid drainage and extracellular space and underlying mechanisms were investigated. When compared to intravenous anesthetic dexmedetomidine or pentobarbital sodium, inhalational isoflurane induced a restricted diffusion of extracellular space, a decreased extracellular space volume fraction, and an increased norepinephrine level in the caudate nucleus or thalamus with the slowdown of brain interstitial fluid drainage. A local administration of norepinephrine receptor antagonists, propranolol, atipamezole and prazosin into extracellular space increased diffusion of extracellular space and interstitial fluid drainage whilst norepinephrine decreased diffusion of extracellular space and interstitial fluid drainage. These findings suggested that restricted diffusion in brain extracellular space can cause slowdown of interstitial fluid drainage, which may contribute to the neurotoxicity following the waste accumulation in extracellular space under inhaled anesthesia per se.


Anesthetics, Inhalation/administration & dosage , Dexmedetomidine/administration & dosage , Extracellular Fluid/drug effects , Extracellular Space/drug effects , Hypnotics and Sedatives/administration & dosage , Isoflurane/administration & dosage , Pentobarbital/administration & dosage , Administration, Inhalation , Administration, Intravenous , Animals , Biological Transport , Brain , Caudate Nucleus/metabolism , Drainage , Humans , Imidazoles/administration & dosage , Male , Norepinephrine/metabolism , Prazosin/administration & dosage , Propranolol/administration & dosage , Rats, Sprague-Dawley , Thalamus/metabolism
...