Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
1.
Int J Pharm ; 654: 123983, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38460768

ABSTRACT

AIM: The study aimed to develop enzyme-degradable nanoparticles comprising polyphosphates and metal cations providing sustained release of the antibacterial drug ethacridine (ETH). METHODS: Calcium polyphosphate (Ca-PP), zinc polyphosphate (Zn-PP) and iron polyphosphate nanoparticles (Fe-PP NPs) were prepared by co-precipitation of sodium polyphosphate with cations and ETH. Developed nanocarriers were characterized regarding particle size, PDI, zeta potential, encapsulation efficiency and drug loading. Toxicological profile of nanocarriers was assessed via hemolysis assay and cell viability on human blood erythrocytes and HEK-293 cells, respectively. The enzymatic degradation of NPs was evaluated in presence of alkaline phosphatase (ALP) monitoring the release of monophosphate, shift in zeta potential and particle size as well as drug release. The antibacterial efficacy against Escherichia coli was determined via microdilution assay. RESULTS: NPs were obtained in a size range between 300 - 480 nm displaying negative zeta potential values. Encapsulation efficiency was in the range of 83.73 %- 95.99 %. Hemolysis assay underlined sufficient compatibility of NPs with blood cells, whereas drug and NPs showed a concentration dependent effect on HEK-293 cells viability. Ca- and Zn-PP NPs exhibited remarkable changes in zeta potential, particle size, monophosphate and drug release upon incubation with ALP, compared to Fe-PP NPs showing only minor differences. The released ETH from Ca- and Zn-PP nanocarriers retained the antibacterial activity against E. coli, whereas no antibacterial effect was observed with Fe-PP NPs. CONCLUSION: Polyphosphate nanoparticles cross-linked with divalent cations and ETH hold promise for sustained drug delivery triggered by ALP for parental administration.


Subject(s)
Nanoparticles , Phosphoric Monoester Hydrolases , Humans , Pharmaceutical Preparations , Phosphoric Monoester Hydrolases/pharmacology , Drug Liberation , Hemolysis , Escherichia coli , HEK293 Cells , Anti-Bacterial Agents/pharmacology , Cations , Polyphosphates , Particle Size , Drug Carriers/pharmacology
2.
Int Immunopharmacol ; 126: 111315, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38043267

ABSTRACT

OBJECTIVES: To investigate the role of protein tyrosine phosphatase non-receptor type 1 (PTPN1) in mitophagy during sepsis and its underlying mechanisms and determine the therapeutic potential of PTPN1 inhibitors in endotoxemia-induced cardiac dysfunction. METHODS: A mouse model of endotoxemia was established by administering an intraperitoneal injection of lipopolysaccharide (LPS). The therapeutic effect of targeting PTPN1 was evaluated using its inhibitor Claramine (CLA). Mitochondrial structure and function as well as the expression of mitophagy-related proteins were evaluated. Rat H9c2 cardiomyocytes were exposed to mouse RAW264.7 macrophage-derived conditioned medium. Cryptotanshinone, a specific p-STAT3 (Y705) inhibitor, was used to confirm the role of STAT3 in PTPN1-mediated mitophagy following LPS exposure. Electrophoretic mobility shift and dual luciferase reporter assays were performed to discern the mechanisms by which STAT3 regulated the expression of PINK1 and PRKN. RESULTS: CLA alleviated LPS-induced myocardial damage, cardiac dysfunction, and mitochondrial injury and dysfunction in the mouse heart. PTPN1 upregulation exacerbated LPS-induced mitochondrial injury and dysfunction in H9c2 cardiomyocytes, but inhibited LPS-induced mitophagy. LPS promoted the interaction between PTPN1 and STAT3 and reduced STAT3 phosphorylation at Tyr705 (Y705), which was required to inhibit mitophagy by PTPN1. Upon LPS stimulation, PTPN1 negatively regulated the transcription of PINK1 and PRKN through dephosphorylation of STAT3 at Y705. STAT3 regulated the transcription of PINK1 and PRKN by binding to STAT3-responsive elements in their promoters. CONCLUSION: PTPN1 upregulation aggravates endotoxemia-induced cardiac dysfunction by impeding mitophagy through dephosphorylation of STAT3 at Y705 and negative regulation of PINK1 and PRKN transcription.


Subject(s)
Endotoxemia , Heart Diseases , Animals , Mice , Rats , Heart Diseases/metabolism , Lipopolysaccharides/pharmacology , Mitophagy , Myocytes, Cardiac/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
3.
Int Immunopharmacol ; 126: 111278, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38011768

ABSTRACT

Cognitive deficit is one of the challenging complications of type 2 diabetes. Sphingosine 1- phosphate receptors (S1PRs) have been implicated in various neurodegenerative and metabolic disorders. The association of S1PRs and cognition in type 2 diabetes remains elusive. Microglia-mediated neuronal damage could be the thread propagating cognitive deficit. The effects of S1PR2 inhibition on cognition in high-fat diet and streptozotocin-induced diabetic mice were examined in this work. We further assessed microglial activation and putative microglial polarisation routes. Cognitive function loss was observed after four months of diabetes induction in Type 2 diabetes animal model. JTE013, an S1PR2 inhibitor, was used to assess neuroprotection against cognitive decline and neuroinflammation in vitro and in vivo diabetes model. JTE013 (10 mg/kg) improved synaptic plasticity by upregulating psd95 and synaptophysin while reducing cognitive decline and neuroinflammation. It further enhanced anti-inflammatory microglia in the hippocampus and prefrontal cortex (PFC), as evidenced by increased Arg-1, CD206, and YM-1 levels and decreased iNOS, CD16, and MHCII levels. TIGAR, TP53-induced glycolysis and apoptosis regulator, might facilitate the anti-inflammatory microglial phenotype by promoting oxidative phosphorylation and decreasing apoptosis. However, since p53 is a TIGAR suppressor, inhibiting p53 could be beneficial. S1PR2 inhibition increased p-Akt and TIGAR levels and reduced the levels of p53 in the PFC and hippocampus of type 2 diabetic mice, thereby decreasing apoptosis. In vitro, palmitate was used to imitate sphingolipid dysregulation in BV2 cells, followed by conditioned media exposure to Neuro2A cells. JTE013 rescued the palmitate-induced neuronal apoptosis by promoting the anti-inflammatory microglia. In the present study, we demonstrate that the inhibition of S1PR2 improves cognitive function and skews microglia toward anti-inflammatory phenotype in type 2 diabetic mice, thereby promising to be a potential therapy for neuroinflammation.


Subject(s)
Cognitive Dysfunction , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Microglia , Neuroinflammatory Diseases , Palmitates/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism
4.
J Transl Med ; 21(1): 819, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37974250

ABSTRACT

BACKGROUND: The protein tyrosine phosphatase H receptor (PTPRH) is known to regulate the occurrence and development of pancreatic and colorectal cancer. However, its association with glycolysis in non-small cell lung cancer (NSCLC) is still unclear. In this study, we aimed to investigate the relationship between PTPRH expression and glucose metabolism and the underlying mechanism of action. METHODS: The expression of PTPRH in NSCLC cells was evaluated by IHC staining, qRT‒PCR and Western blotting. The effect of PTPRH on cell biological behavior was evaluated by colony assays, EdU experiments, Transwell assays, wound healing assays and flow cytometry. Changes in F-18-fluorodeoxyglucose (18F-FDG) uptake and glucose metabolite levels after altering PTPRH expression were detected via a gamma counter and lactic acid tests. The expression of glycolysis-related proteins in NSCLC cells was detected by Western blotting after altering PTPRH expression. RESULTS: The results showed that PTPRH was highly expressed in clinical patient tissue samples and closely related to tumor diameter and clinical stage. In addition, PTPRH expression was associated with glycometabolism indexes on 18F-FDG positron emission tomography/computed tomography (PET/CT) imaging, the expression level of Ki67 and the expression levels of glycolysis-related proteins. PTPRH altered cell behavior, inhibited apoptosis, and promoted 18F-FDG uptake, lactate production, and the expression of glycolysis-related proteins. In addition, PTPRH modulated the glycometabolism of NSCLC cells via the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway, as assessed using LY294002 and 740Y-P (an inhibitor and agonist of PI3K, respectively). The same results were validated in vivo using a xenograft tumor model in nude mice. Protein expression levels of PTPRH, glycolysis-related proteins, p-PI3K/PI3K and p-AKT/AKT were measured by IHC staining using a subcutaneous xenograft model in nude mice. CONCLUSIONS: In summary, we report that PTPRH promotes glycolysis, proliferation, migration, and invasion via the PI3K/AKT/mTOR signaling pathway in NSCLC and ultimately promotes tumor progression, which can be regulated by LY294002 and 740Y-P. These results suggest that PTPRH is a potential therapeutic target for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Mice , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice, Nude , Lung Neoplasms/pathology , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Phosphoric Monoester Hydrolases/therapeutic use , Fluorodeoxyglucose F18 , Positron Emission Tomography Computed Tomography , Cell Proliferation , Cell Line, Tumor , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Glycolysis , Mammals/metabolism
5.
Article in English | MEDLINE | ID: mdl-37741603

ABSTRACT

We used the gill (Na+, K+)-ATPase as a molecular marker to provide a comprehensive kinetic analysis of the effects of Co2+in vitro on the modulation of K+-phosphatase activity in the Blue crab Callinectes danae. Co2+ can stimulate or inhibit K+-phosphatase activity. With Mg2+, K+-phosphatase activity is almost completely inhibited by Co2+. Co2+ stimulates K+-phosphatase activity similarly to Mg2+ although with a ≈4.5-fold greater affinity. At saturating Mg2+ concentrations, Mg2+ displaces bound Co2+ from the Mg2+-binding site in a concentration dependent manner, but Co2+ cannot displace Mg2+ from its binding site even at millimolar concentrations. Saturation by Co2+ of the Mg2+ binding site does not affect pNPP recognition by the enzyme. Substitution of Mg2+ by Co2+ slightly increases enzyme affinity for K+ and NH4+. Independently of Mg2+, inhibition by ouabain or sodium ions is unaffected by Co2+. Investigation of gill (Na+, K+)-ATPase K+-phosphatase activity provides a reliable tool to examine the kinetic effects of Co2+ with and without Na+ and ATP. Given that the toxic effects of Co2+ at the molecular level are poorly understood, these findings advance our knowledge of the mechanism of action of Co2+ on the crustacean gill (Na+, K+)-ATPase.


Subject(s)
Brachyura , Animals , Sodium-Potassium-Exchanging ATPase/metabolism , Kinetics , Cobalt/toxicity , Gills/metabolism , Ions , Sodium/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology
6.
J Environ Manage ; 342: 118207, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37263035

ABSTRACT

Nitrogen (N) and phosphorus (P) are two important nutrient elements that limit the growth of plants and microorganisms. The effect of the N supply on soil P cycling and its mechanism remain poorly known. Here, we characterized the effects of different N application rates on soil P availability, the abundances of P-cycling functional genes, and microbial communities involved in P-cycling following the application of N for 13 years in a tea plantation. Soil available P (AP) decreased significantly under N application. The opposite pattern was observed for the activity of soil phosphatases including alkaline (ALP) and acid phosphatase (ACP). Furthermore, N addition increased the abundance of ppa but decreased the abundance of phoD in soil. Both ppa- and phoD-harboring communities varied with N application levels. Redundancy analysis (RDA) showed that soil pH was a key variable modulating ppa-harboring and phoD-harboring microbial communities. Partial least squares path modeling (PLS-PM) revealed that long-term N application indirectly reduced soil P availability by altering the abundances of phoD-harboring biomarker taxa. Overall, our findings indicated that N-induced reductions in AP increased microbial competition for P by selecting microbes with P uptake and starvation response genes or those with phosphatases in tea plantation system. This suggests that tea plantations should be periodically supplemented with P under N application, especially under high N application levels.


Subject(s)
Camellia sinensis , Microbiota , Soil/chemistry , Phosphorus/analysis , Nitrogen/analysis , Soil Microbiology , Phosphoric Monoester Hydrolases/pharmacology , Tea
7.
Am J Physiol Cell Physiol ; 324(6): C1320-C1331, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37154493

ABSTRACT

Radiation-induced heart disease (RIHD) progresses over time and may manifest decades after the initial radiation exposure, which is associated with significant morbidity and mortality. The clinical benefit of radiotherapy is always counterbalanced by an increased risk of cardiovascular events in survivors. There is an urgent need to explore the effect and the underlying mechanism of radiation-induced heart injury. Mitochondrial damage widely occurs in irradiation-induced injury, and mitochondrial dysfunction contributes to necroptosis development. Experiments were performed using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and rat H9C2 cells to investigate the effect of mitochondrial injury on necroptosis in irradiated cardiomyocytes and to further elucidate the mechanism underlying radiation-induced heart disease and discover possible preventive targets. After γ-ray irradiation, the expression levels of necroptosis markers were increased, along with higher oxidative stress and mitochondrial injury. These effects could be abated by overexpression of protein tyrosine phosphatase, mitochondrial 1 (PTPMT1). Inhibiting oxidative stress or increasing the expression of PTPMT1 could protect against radiation-induced mitochondrial injury and then decrease the necroptosis of cardiomyocytes. These results suggest that PTPMT1 may be a new target for the treatment of radiation-induced heart disease.NEW & NOTEWORTHY Effective strategies are still lacking for treating RIHD, with unclear pathological mechanisms. In cardiomyocytes model of radiation-induced injuries, we found γ-ray irradiation decreased the expression of PTPMT1, increased oxidative stress, and induced mitochondrial dysfunction and necroptosis in iPSC-CMs. ROS inhibition attenuated radiation-induced mitochondrial damage and necroptosis. PTPMT1 protected cardiomyocytes from necroptosis induced by γ-ray irradiation by alleviating mitochondrial injury. Therefore, PTPMT1 might be a potential strategy for treating RIHD.


Subject(s)
Heart Diseases , Myocytes, Cardiac , Animals , Rats , Heart Diseases/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Necroptosis , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology
8.
Antiviral Res ; 213: 105601, 2023 05.
Article in English | MEDLINE | ID: mdl-37068596

ABSTRACT

Phosphatidylinositol lipids play vital roles in lipid signal transduction, membrane recognition, vesicle transport, and viral replication. Previous studies have revealed that SAC1-like phosphatidylinositol phosphatase (SACM1L/SAC1), which uses phosphatidylinositol-4-phosphate (PI4P) as its substrate, greatly affects the replication of certain bacteria and viruses in vitro. However, it remains unclear whether and how SAC1 modulates hepatitis B virus (HBV) replication in vitro and in vivo. In the present study, we observed that SAC1 silencing significantly increased HBV DNA replication, subviral particle (SVP) expression, and secretion of HBV virions, whereas SAC1 overexpression exerted the opposite effects. Moreover, SAC1 overexpression inhibited HBV DNA replication and SVP expression in a hydrodynamic injection-based HBV-persistent replicating mouse model. Mechanistically, SAC1 silencing increased the number of HBV-containing autophagosomes as well as PI4P levels on the autophagosome membrane. Moreover, SAC1 silencing blocked autophagosome-lysosome fusion by inhibiting the interaction between synaptosomal-associated protein 29 and vesicle-associated membrane protein 8. Collectively, our data indicate that SAC1 significantly inhibits HBV replication by promoting the autophagic degradation of HBV virions. Our findings support that SAC1-mediated phospholipid metabolism greatly modulates certain steps of the HBV life-cycle and provide a new theoretical basis for antiviral therapy.


Subject(s)
Hepatitis B virus , Hepatitis B , Animals , Mice , Hepatitis B virus/genetics , Virus Replication , Hepatitis B/metabolism , Phosphatidylinositols/pharmacology , Virion/metabolism , Phosphoric Monoester Hydrolases/pharmacology
9.
Cell Mol Neurobiol ; 43(6): 2989-3003, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37106272

ABSTRACT

Elabela (ELA), which is the second endogenous peptide ligand of the apelin receptor (APJ) to be discovered, has been widely studied for potential use as a therapeutic peptide. However, its role in ischemic stroke (IS), which is a leading cause of disability and death worldwide and has limited therapeutic options, is uncertain. The aim of the present study was to investigate the beneficial effects of ELA on neuron survival after ischemia and the underlying molecular mechanisms. Primary cortical neurons were isolated from the cerebral cortex of pregnant C57BL/6J mice. Flow cytometry and immunofluorescence showed that ELA inhibited oxygen-glucose deprivation (OGD) -induced apoptosis and axonal damage in vitro. Additionally, analysis of the Gene Expression Omnibus database revealed that the expression of microRNA-124-3p (miR-124-3p) was decreased in blood samples from patients with IS, while the expression of C-terminal domain small phosphatase 1 (CTDSP1) was increased. These results indicated that miR-124-3p and CTDSP1 were related to ischemic stroke, and there might be a negative regulatory relationship between them. Then, we found that ELA significantly elevated miR-124-3p expression, suppressed CTDSP1 expression, and increased p-AKT expression by binding to the APJ receptor under OGD in vitro. A dual-luciferase reporter assay confirmed that CTDSP1 was a direct target of miR-124-3p. Furthermore, adenovirus-mediated overexpression of CTDSP1 exacerbated neuronal apoptosis and axonal damage and suppressed AKT phosphorylation, while treatment with ELA or miR-124-3p mimics reversed these effects. In conclusion, these results indicated that ELA could alleviate neuronal apoptosis and axonal damage by upregulating miR-124-3p and activating the CTDSP1/AKT signaling pathway. This study, for the first time, verified the protective effect of ELA against neuronal injury after ischemia and revealed the underlying mechanisms. We demonstrated the potential for the use of ELA as a therapeutic agent in the treatment of ischemic stroke.


Subject(s)
Ischemic Stroke , MicroRNAs , Neuroprotective Agents , Mice , Animals , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-akt , Phosphoric Monoester Hydrolases/pharmacology , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Peptides/pharmacology , Apoptosis , Glucose/metabolism
10.
Behav Brain Res ; 446: 114415, 2023 05 28.
Article in English | MEDLINE | ID: mdl-36997095

ABSTRACT

Sphingosine-1-phosphate (S1P) is emerging as a crucial sphingolipid modulating neuroinflammation and cognition. S1P levels in the brain have been found to be decreased in cognitive impairment. S1P lyase (S1PL) is the key enzyme in metabolizing S1P and has been implicated in neuroinflammation. This study evaluated the effect of S1PL inhibition on cognition in type 2 diabetic mice. Fingolimod (0.5 mg/kg and 1 mg/kg) rescued cognition in high-fat diet and streptozotocin-induced diabetic mice, as evident in the Y maze and passive avoidance test. We further evaluated the effect of fingolimod on the activation of microglia in the pre-frontal cortex (PFC) and hippocampus of diabetic mice. Our study revealed that fingolimod inhibited S1PL and promoted anti-inflammatory microglia in both PFC and hippocampus of diabetic mice as it increased Ym-1 and arginase-1. The levels of p53 and apoptotic proteins (Bax and caspase-3) were elevated in the PFC and hippocampus of type 2 diabetic mice which fingolimod reversed. The underlying mechanism promoting anti-inflammatory microglial phenotype was also explored in this study. TIGAR, TP53-associated glycolysis and apoptosis regulator, is known to foster anti-inflammatory microglia and was found to be downregulated in the brain of type 2 diabetic mice. S1PL inhibition decreased the levels of p53 and promoted TIGAR, thereby increasing anti-inflammatory microglial phenotype and inhibiting apoptosis in the brain of diabetic mice. Our study reveals that S1PL inhibition could be beneficial in mitigating cognitive deficits in diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Mice , Animals , Sphingosine/pharmacology , Sphingosine/metabolism , Fingolimod Hydrochloride/metabolism , Fingolimod Hydrochloride/pharmacology , Microglia , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Neuroinflammatory Diseases , Cognition , Diabetes Mellitus, Type 2/metabolism , Phosphates/metabolism , Phosphates/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Apoptosis Regulatory Proteins/metabolism
11.
ACS Appl Bio Mater ; 6(1): 164-170, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36525564

ABSTRACT

Drug-resistant breast cancers such as Triple negative breast cancer (TNBC) do not respond successfully to chemotherapy treatments because they lack the expression of receptor targets. Drug-resistant anti-cancer treatments require innovative approaches to target these cells without relying on the receptors. Intracellular self-assembly of small molecules induced by enzymes is a nanotechnology approach for inhibiting cancer cell growth. In this approach, enzymes will induce the self-assembly of small molecules to nanofibers, which leads to cell death. Here, we investigate the self-assembly of a modified small peptide induced by two different phosphatases: alkaline phosphatase (ALP) and eye absent tyrosine phosphatase (EYA). ALPs are expressed in many adult human tissues and are critical for many cellular functions. EYAs are embryonic enzymes that are over-expressed in drug-resistant breast cancers. We synthesized a small diphenylalanine-based peptide with a tyrosine phosphate end group as the substrate of phosphatase enzymes. Peptides were synthesized with solid phase techniques and were characterized by HPLC and MALDI-TOF. To characterize the self-assembly of peptides exposed to enzymes, different techniques were used such as scattering light intensity, microscopes, and phosphate detection kit. We then determined the toxicity effect of the peptide against normal breast cancer cells, MCF-7, and drug-resistant breast cancer cells, MDA-MB-231. The results showed that the EYA enzyme is able to initiate self-assembly at lower peptide concentration with higher self-assembling intensity compared to ALP. A significant decrease in the TNBC cell number was observed even with a low peptide concentration of 60 µM. These results collectively support the exploration of enzyme self-assembly to treat TNBC.


Subject(s)
Nanofibers , Triple Negative Breast Neoplasms , Humans , Alkaline Phosphatase , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Nanofibers/chemistry , Peptides/pharmacology , Phosphoric Monoester Hydrolases/pharmacology , Phosphoric Monoester Hydrolases/therapeutic use , Cell Proliferation
12.
Article in English | MEDLINE | ID: mdl-36232061

ABSTRACT

Microcapsules serve as a feasible formulation to load phenolic substances such as salicylic acid, a natural and safe antimicrobial agent. However, the antibacterial efficacy of salicylic acid microcapsules (SAMs) remains to be elucidated. Here, salicylic acid/ß-cyclodextrin inclusion microcapsules were subjected to systematic antibacterial assays and preliminary antibacterial mechanism tests using Escherichia coli and Staphylococcus aureus as target organisms. It was found that the core-shell rhomboid-shaped SAMs had a smooth surface. SAMs exhibited a minimum inhibitory concentration (MIC) and a minimum bactericidal concentration (MBC) of 4 mg/mL against both bacteria. In the growth inhibition assay, 1/4 × MIC, 1/2 × MIC, and 1 × MIC of SAMs effectively retarded bacterial growth, and this effect was more prominent with the rise in the level of SAMs. Practically, SAMs possessed a rapid bactericidal effect at the 1 × MIC level with a reduction of more than 99.9% bacterial population within 10 min. A pronounced sterilization activity against E. coli and S. aureus was also observed when SAMs were embedded into hand sanitizers as antimicrobial agents. Moreover, exposure of both bacteria to SAMs resulted in the leakage of intracellular alkaline phosphatases and macromolecular substances (nucleic acids and proteins), which indicated the disruption of bacterial cell walls and cell membranes. In conclusion, SAMs were able to inactivate E. coli and S. aureus both in vitro and in situ, highlighting the promising utilization of this formulation for antimicrobial purposes in the area of food safety and public health.


Subject(s)
Hand Sanitizers , Nucleic Acids , beta-Cyclodextrins , Anti-Bacterial Agents/pharmacology , Bacteria , Capsules/pharmacology , Escherichia coli , Hand Sanitizers/pharmacology , Microbial Sensitivity Tests , Phosphoric Monoester Hydrolases/pharmacology , Salicylic Acid/pharmacology , Staphylococcus aureus
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 42(9): 1303-1308, 2022 Sep 20.
Article in Chinese | MEDLINE | ID: mdl-36210702

ABSTRACT

OBJECTIVE: To investigate the regulatory role of miRNA-26a in vascular smooth muscle cell (VSMC) calcification by regulating connective tissue growth factor (CTGF). METHODS: Rat thoracic aorta VSMCs (A7r5 cells) with induced calcification were treated with AR234960 agonist or transfected with miR-26a mimic, or with both treatments. Alizarin red staining was used to determine calcium deposition, and phosphatase (ALP) activity in the cells was measured. The mRNA and protein expressions of miR-26a, OPG, OPN, BMP-2 and collagen Ⅱ were detected using qPCR and Western blotting. The binding of miR-26a to CTGF was verified using dual luciferase reporter gene assay. RESULTS: After induced calcification, A7r5 cells showed gradually decreased miR-26a expression (P < 0.05) and progressively increased CTGF expression (P < 0.05) with the extension of induction time. Treatment of the cells with AR234960 obviously increased calcification in the cells, while transfection with miR-26a mimic significantly reduced cell calcification. The calcifying cells showed significantly increased ALP activity and expressions of OPN, BMP-2 and collagen Ⅱ (P < 0.05) and lowered OPG expression (P < 0.05), and treatment with AR234960 did not produce obvious effects on these changes (P > 0.05). Transfection with miR-26a mimic resulted in significantly decreased ALP activity and expressions OPN, BMP-2 and collagen Ⅱ expression (P < 0.05) and increased OPG expression (P < 0.05) in the calcifying cells. These effects of miR-26a mimic was significantly attenuated by treatment of the cells with AR234960 (P < 0.05). The result of luciferase reporter gene assay confirmed the binding of miR-26a to CTGF. CONCLUSION: miRNA-26a can effectively alleviate vascular calcification by lowering the level of CTGF, reducing ALP activity and the expressions of OPN, BMP-2 and collagen Ⅱ, and increasing the expression of OPG.


Subject(s)
MicroRNAs , Vascular Calcification , Animals , Calcium/metabolism , Cells, Cultured , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/pharmacology , MicroRNAs/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , RNA, Messenger/metabolism , Rats , Sulfones
14.
Mol Med ; 28(1): 125, 2022 10 22.
Article in English | MEDLINE | ID: mdl-36273174

ABSTRACT

BACKGROUND: Oxidative stress-caused damage to the retinal pigment epithelium (RPE) underlies the onset and progression of age-related macular degeneration (AMD). Impaired mitochondrial biogenesis sensitizes RPE cells to mitochondrial dysfunction, energy insufficiency and death. Src-homology 2 domain-containing phosphatase (SHP)-1 is important in regulating immune responses and cell survival. However, its roles in cell survival are not always consistent. Until now, the effects of SHP-1 on RPE dysfunction, especially mitochondrial homeostasis, remain to be elucidated. We sought to clarify the effects of SHP-1 in RPE cells in response to atRAL-induced oxidative stress and determine the regulatory mechanisms involved. METHODS: In the all trans retinal (atRAL)-induced oxidative stress model, we used the vector of lentivirus to knockdown the expression of SHP-1 in ARPE-19 cells. CCK-8 assay, Annexin V/PI staining and JC-1 staining were utilized to determine the cell viability, cell apoptosis and mitochondrial membrane potential. We also used immunoprecipitation to examine the ubiquitination modification of stimulator of interferon genes (STING) and its interaction with SHP-1. The expression levels of mitochondrial marker, proteins related to mitochondrial biogenesis, and signaling molecules involved were examined by western blotting analysis. RESULTS: We found that SHP-1 knockdown predisposed RPE cells to apoptosis, aggravated mitochondrial damage, and repressed mitochondrial biogenesis after treatment with atRAL. Immunofluoresent staining and immunoprecipitation analysis confirmed that SHP-1 interacted with the endoplasmic reticulum-resident STING and suppressed K63-linked ubiquitination and activation of STING. Inhibition of STING with the specific antagonist H151 attenuated the effects of SHP-1 knockdown on mitochondrial biogenesis and oxidative damage. The adenosine monophosphate-activated protein kinase (AMPK) pathway acted as the crucial downstream target of STING and was involved in the regulatory processes. CONCLUSIONS: These findings suggest that SHP-1 knockdown potentiates STING overactivation and represses mitochondrial biogenesis and cell survival, at least in part by blocking the AMPK pathway in RPE cells. Therefore, restoring mitochondrial health by regulating SHP-1 in RPE cells may be a potential therapeutic strategy for degenerative retinal diseases including AMD.


Subject(s)
Macular Degeneration , Mitochondria , Retinal Pigment Epithelium , Retinaldehyde , Humans , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , AMP-Activated Protein Kinases/metabolism , Annexin A5/metabolism , Annexin A5/pharmacology , Apoptosis/genetics , Interferons/genetics , Interferons/metabolism , Interferons/pharmacology , Macular Degeneration/genetics , Macular Degeneration/metabolism , Mitochondria/metabolism , Organelle Biogenesis , Oxidative Stress , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retinaldehyde/metabolism , Retinaldehyde/pharmacology
15.
Environ Monit Assess ; 194(10): 718, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36050604

ABSTRACT

Microplastics (MPs) are critical emerging pollutants around the world. There is a growing interest in the effects of MP ingestion, non-digestion, and toxicity on aquatic organisms. Amphibian tadpoles are the vertebrate group that has received the least attention regarding this issue. The aim of the present study was to determine the ingestion of polyethylene MPs by Scinax squalirostris tadpoles by atomic force microscopy (AFM) and to evaluate the activities of carboxylesterase (CbE, using 4-naphthyl butyrate-NB-, and 1-naphthyl acetate -NA- as substrates) and alkaline phosphatase (ALP) under MP exposure. Enzyme activities were analyzed spectrophotometrically at 2 and 10 days of exposure. Tadpoles were exposed to two different treatments during 10 days: a negative control (CO, dechlorinated water) and MP (60 mg L-1). AFM images of the digestive contents of tadpoles revealed the presence of MPs. After 10 days of MP exposure, CbE (NB) activity was significantly higher and CbE (NA) activity was significantly lower in MP treatments than in controls. ALP activity decreased in MP treatments after 2 and 10 days of exposure. The detection of MP particles in the intestinal contents and the effects on metabolic enzymes in a common frog species evidenced the potential health risk of MP to aquatic vertebrates. Thus, the differential response in enzymes and substrates demonstrate the need for considering the complex effects of contaminants and nutrients on ecosystems for ecotoxicological risk characterization.


Subject(s)
Microplastics , Water Pollutants, Chemical , Animals , Anura , Carboxylesterase/pharmacology , Ecosystem , Environmental Monitoring , Larva , Phosphoric Monoester Hydrolases/pharmacology , Plastics , Water Pollutants, Chemical/toxicity
16.
Microbiol Spectr ; 10(5): e0051922, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36094204

ABSTRACT

Aspergillus fumigatus is a deadly opportunistic fungal pathogen responsible for ~100,000 annual deaths. Azoles are the first line antifungal agent used against A. fumigatus, but azole resistance has rapidly evolved making treatment challenging. Caspofungin is an important second-line therapy against invasive pulmonary aspergillosis, a severe A. fumigatus infection. Caspofungin functions by inhibiting ß-1,3-glucan synthesis, a primary and essential component of the fungal cell wall. A phenomenon termed the caspofungin paradoxical effect (CPE) has been observed in several fungal species where at higher concentrations of caspofungin, chitin replaces ß-1,3-glucan, morphology returns to normal, and growth rate increases. CPE appears to occur in vivo, and it is therefore clinically important to better understand the genetic contributors to CPE. We applied genomewide association (GWA) analysis and molecular genetics to identify and validate candidate genes involved in CPE. We quantified CPE across 67 clinical isolates and conducted three independent GWA analyses to identify genetic variants associated with CPE. We identified 48 single nucleotide polymorphisms (SNPs) associated with CPE. We used a CRISPR/Cas9 approach to generate gene deletion mutants for seven genes harboring candidate SNPs. Two null mutants, ΔAfu3g13230 and ΔAfu4g07080 (dscP), resulted in reduced basal growth rate and a loss of CPE. We further characterized the dscP phosphatase-null mutant and observed a significant reduction in conidia production and extremely high sensitivity to caspofungin at both low and high concentrations. Collectively, our work reveals the contribution of Afu3g13230 and dscP in CPE and sheds new light on the complex genetic interactions governing this phenotype. IMPORTANCE This is one of the first studies to apply genomewide association (GWA) analysis to identify genes involved in an Aspergillus fumigatus phenotype. A. fumigatus is an opportunistic fungal pathogen that causes hundreds of thousands of infections and ~100,000 deaths each year, and antifungal resistance has rapidly evolved in this species. A phenomenon called the caspofungin paradoxical effect (CPE) occurs in some isolates, where high concentrations of the drug lead to increased growth rate. There is clinical relevance in understanding the genetic basis of this phenotype, since caspofungin concentrations could lead to unintended adverse clinical outcomes in certain cases. Using GWA analysis, we identified several interesting candidate polymorphisms and genes and then generated gene deletion mutants to determine whether these genes were important for CPE. Two of these mutant strains (ΔAfu3g13230 and ΔAfu4g07080/ΔdscP) displayed a loss of the CPE. This study sheds light on the genes involved in clinically important phenotype CPE.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Caspofungin/pharmacology , Caspofungin/metabolism , Aspergillus fumigatus/genetics , Antifungal Agents/pharmacology , Echinocandins/pharmacology , Echinocandins/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Azoles/metabolism , Azoles/pharmacology , Chitin , Genomics , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology
17.
Pestic Biochem Physiol ; 187: 105199, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36127070

ABSTRACT

Biocontrol microbes are environment-friendly and safe for humans and animals. To seek biocontrol microbes effective in suppressing tomato gray mold is important for tomato production. Therefore, serial experiments were conducted to characterize the antagonism of Bacillus velezensis HY19, a novel self-isolated biocontrol bacterium, against Botrytis cinerea in vitro and the control on tomato gray mold in greenhouse. This bacterium produced extracellular phosphatase, protease, cellulose and siderophores, and considerably inhibited the growth of B. cinerea. A liquid chromatography-mass spectrometry (LC-MS) detected salicylic acid and numerous antifungal substances present in B. velezensis HY19 fermentation liquid (BVFL). When B. cinerea was grown on potato glucose agar, BVFL crude extract remarkably suppressed the fungal growth and reduced protein content and the activities of catalase (CAT), superoxide dismutase (SOD), and peroxidase (POD). Transcriptome studies showed that BVFL crude extract significantly induced different expression of numerous genes in B. cinerea, most of which were down-regulated. Theses differently expressed genes were involved in the biological process, cell compartment, molecular functions, and metabolisms of glycine, serine, threonine, and sulfur in pathogen hyphae. Thus, this biocontrol bacterium antagonized B. cinerea in multiple ways due to the production of numerous antifungal substances that acted on multiple targets in the cells. BVFL significantly increased antioxidant enzyme activities in tomato leaves and decreased the incidence of tomato gray mold, with the control efficacies of 73.12-76.51%. Taken together, B. velezensis HY19 showed a promising use potential as a powerful bioagent against tomato gray mold.


Subject(s)
Solanum lycopersicum , Agar/pharmacology , Antifungal Agents/pharmacology , Antioxidants/pharmacology , Bacillus , Catalase , Cellulose/pharmacology , Complex Mixtures/pharmacology , Glucose/pharmacology , Glycine/pharmacology , Solanum lycopersicum/microbiology , Peptide Hydrolases/pharmacology , Phosphoric Monoester Hydrolases/pharmacology , Plant Diseases/microbiology , Plant Diseases/prevention & control , Salicylic Acid/pharmacology , Serine/pharmacology , Siderophores/pharmacology , Sulfur/pharmacology , Superoxide Dismutase , Threonine/pharmacology
18.
J Hypertens ; 40(12): 2502-2512, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36093879

ABSTRACT

BACKGROUND: Hypertension is a severe public health risk factor worldwide. Elevated angiotensin II (Ang II) produced by the renin-angiotensin-aldosterone system can lead to hypertension and its complications. METHOD: In this study, we addressed the cardiac-injury effects of Ang II and investigated the signaling mechanism induced by Ang II. Both H9c2 cardiomyoblast cells and neonatal rat cardiomyocytes were exposed to Ang II to observe hypertension-related cardiac apoptosis. RESULTS: The results of western blotting revealed that Ang II significantly attenuated the IGF1R-PI3K-AKT pathway via the Ang II-AT1 receptor axis and phosphatase and tensin homolog expression. Furthermore, real-time PCR showed that Ang II also activated miR-320-3p transcription to repress the PI3K-Akt pathway. In the heart tissue of spontaneously hypertensive rats, activation of the IGF1R survival pathway was also reduced compared with that in Wistar-Kyoto rats, especially in aged spontaneously hypertensive rats. CONCLUSION: Hence, we speculate that the Ang II-AT1 receptor axis induces both phosphatase and tensin homolog and miR-320-3p expression to downregulate the IGF1R-PI3K-AKT survival pathway and cause cell apoptosis in the heart.


Subject(s)
Hypertension , MicroRNAs , Rats , Animals , Angiotensin II/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Angiotensin, Type 1/metabolism , Tensins/metabolism , Rats, Inbred SHR , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Rats, Inbred WKY , Apoptosis , Myocytes, Cardiac/metabolism , Hypertension/metabolism , MicroRNAs/metabolism
19.
Environ Res ; 215(Pt 3): 114363, 2022 12.
Article in English | MEDLINE | ID: mdl-36174759

ABSTRACT

Glyphosate (N-(phosphonomethyl)glycine) is broad-spectrum herbicide that is extensively used worldwide, but its effects on the soil microbiome are inconsistent. To provide a sound scientific basis for herbicide re-review and registration decisions, we conducted a four-year (2013-2016) study in which we consecutively applied glyphosate to a wheat (Triticum aestivum L.)-field pea (Pisum sativum L.)-canola (Brassica napus L.)-wheat crop rotation at five sites in the Canadian prairies. The glyphosate rates were 0, 1, 2, 4 and 8 kg ae ha-1, applied pre-seeding and post-harvest every year. The wheat rhizosphere was sampled in the final year of the study and analysed for microbial biomass C (MBC), the composition and diversity of the microbiome, and activities of ß-glucosidase, N-acetyl-ß-glucosiminidase, acid phosphomonoesterase and arylsulphatase. Glyphosate did not affect MBC, the composition and diversity of prokaryotes and fungi, and the activities of three of the four enzymes measured in the wheat rhizosphere. The one effect of glyphosate was a wave-like response of N-acetyl-ß-glucosaminidase activity with increasing application rates. The experimental sites had much greater effects, driven by soil pH and organic C, on the soil microbiome composition and enzyme activities than glyphosate. Soil pH was positively correlated with the relative abundance of Acidobacteriota but negatively correlated with that of Actinobacteriota and Basidiomycota. Soil organic C was positively correlated with the relative abundances of Proteobacteriota and Verrucomicrobiota, but negatively correlated with the relative abundance of Crenachaeota. The activity of acid phosphomonoesterase declined with increasing relative abundance of Acidobacteriota, but increased with that of Actinobacteriota and Basidiomycota. The activity of N-acetyl-ß-glucosaminidase also increased with increasing relative abundance of Actinobacteriota but decreased with that of Mortierellomycota. ß-glucosidase activity also decreased with increasing relative abundance of Mortierellomycota. The core fungal species observed in at least 90% of the samples were Humicola nigrescens, Gibberella tricincta and Giberella fujikuroi. Therefore, this multi-site study on the Canadian prairies revealed no significant effects of 4-year applications of glyphosate applied at different rates on most soil microbial properties despite differences in the properties among sites. However, it is important to keep evaluating glyphosate effects on the soil microbiome and its functioning because it is the most widely used herbicide worldwide.


Subject(s)
Cellulases , Herbicides , Microbiota , Arylsulfatases/pharmacology , Bacteria , Canada , Cellulases/pharmacology , Glycine/analogs & derivatives , Herbicides/toxicity , Hexosaminidases/pharmacology , Phosphoric Monoester Hydrolases/pharmacology , Rhizosphere , Soil/chemistry , Soil Microbiology , Triticum , Glyphosate
20.
J Mol Cell Biol ; 14(7)2022 09 27.
Article in English | MEDLINE | ID: mdl-36002018

ABSTRACT

In mammals, the growth and maturation of oocytes within growing follicles largely depends on ovarian granulosa cells (GCs) in response to gonadotropin stimulation. Many signals have been shown to regulate GC proliferation and apoptosis. However, whether the tyrosine phosphatase SHP2 is involved remains unclear. In this study, we identified the crucial roles of SHP2 in modulating GC proliferation and apoptosis. The production of both mature oocytes and pups was increased in mice with Shp2 specifically deleted in ovarian GCs via Fshr-Cre. Shp2 deletion simultaneously promoted GC proliferation and inhibited GC apoptosis. Furthermore, Shp2 deficiency promoted, while Shp2 overexpression inhibited, the proliferation of cultured primary mouse ovarian GCs and the human ovarian granulosa-like tumor cell line KGN in vitro. Shp2 deficiency promoted follicule-stimulating hormone (FSH)-activated phosphorylation of AKT in vivo. SHP2 deficiency reversed the inhibitory effect of hydrogen peroxide (H2O2) on AKT activation in KGN cells. H2O2 treatment promoted the interaction between SHP2 and the p85 subunit of PI3K in KGN cells. Therefore, SHP2 in GCs may act as a negative modulator to balance follicular development by suppressing PI3K/AKT signaling. The novel function of SHP2 in modulating proliferation and apoptosis of GCs provides a potential therapeutic target for the clinical treatment of follicle developmental dysfunction.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Female , Mice , Humans , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Hydrogen Peroxide/metabolism , Granulosa Cells/metabolism , Tyrosine/metabolism , Tyrosine/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/pharmacology , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL
...