Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 7077, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37127686

ABSTRACT

Alzheimer's disease (AD) is a chronic degenerative brain disorder with no clear pathogenesis or effective cure, accounting for 60-80% of cases of dementia. In recent years, the importance of neuroinflammation in the pathogenesis of AD and other neurodegenerative disorders has come into focus. Previously, we made the serendipitous discovery that the widely used drug excipient N,N-dimethylacetamide (DMA) attenuates endotoxin-induced inflammatory responses in vivo. In the current work, we investigate the effect of DMA on neuroinflammation and its mechanism of action in in-vitro and ex-vivo models of AD. We show that DMA significantly suppresses the production of inflammatory mediators, such as reactive oxygen species (ROS), nitric oxide (NO) and various cytokines and chemokines, as well as amyloid-ß (Aß), in cultured microglia and organotypic hippocampal slices induced by lipopolysaccharide (LPS). We also demonstrate that DMA inhibits Aß-induced inflammation. Finally, we show that the mechanism of DMA's effect on neuroinflammation is inhibition of the nuclear factor kappa-B (NF-κB) signaling pathway and we show how DMA dismantles the positive feedback loop between NF-κB and Aß synthesis. Taken together, our findings suggest that DMA, a generally regarded as safe compound that crosses the blood brain barrier, should be further investigated as a potential therapy for Alzheimer's disease and neuroinflammatory disorders.


Subject(s)
Alzheimer Disease , Humans , NF-kappa B/metabolism , Neuroinflammatory Diseases , Signal Transduction , Amyloid beta-Peptides/metabolism
2.
Signal Transduct Target Ther ; 7(1): 135, 2022 04 23.
Article in English | MEDLINE | ID: mdl-35461318

ABSTRACT

The role of microbiota in health and diseases is being highlighted by numerous studies since its discovery. Depending on the localized regions, microbiota can be classified into gut, oral, respiratory, and skin microbiota. The microbial communities are in symbiosis with the host, contributing to homeostasis and regulating immune function. However, microbiota dysbiosis can lead to dysregulation of bodily functions and diseases including cardiovascular diseases (CVDs), cancers, respiratory diseases, etc. In this review, we discuss the current knowledge of how microbiota links to host health or pathogenesis. We first summarize the research of microbiota in healthy conditions, including the gut-brain axis, colonization resistance and immune modulation. Then, we highlight the pathogenesis of microbiota dysbiosis in disease development and progression, primarily associated with dysregulation of community composition, modulation of host immune response, and induction of chronic inflammation. Finally, we introduce the clinical approaches that utilize microbiota for disease treatment, such as microbiota modulation and fecal microbial transplantation.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Dysbiosis/therapy , Homeostasis , Humans , Immunity , Inflammation
3.
Reprod Sci ; 29(10): 2894-2907, 2022 10.
Article in English | MEDLINE | ID: mdl-35349119

ABSTRACT

Preterm birth accounts for the majority of perinatal mortality worldwide, and there remains no FDA-approved drug to prevent it. Recently, we discovered that the common drug excipient, N,N-dimethylacetamide (DMA), delays inflammation-induced preterm birth in mice by inhibiting NF-κB. Since we reported this finding, it has come to light that a group of widely used, structurally related aprotic solvents, including DMA, N-methyl-2-pyrrolidone (NMP) and dimethylformamide (DMF), have anti-inflammatory efficacy. We show here that DMF suppresses LPS-induced TNFα secretion from RAW 264.7 cells and IL-6 and IL-8 secretion from HTR-8 cells at concentrations that do not significantly affect cell viability. Like DMA, DMF protects IκBα from degradation and prevents the p65 subunit of NF-κB from translocating to the nucleus. In vivo, DMF decreases LPS-induced inflammatory cell infiltration and expression of TNFα and IL-6 in the placental labyrinth, all to near baseline levels. Finally, DMF decreases the rate of preterm birth in LPS-induced pregnant mice (P<.0001) and the rate at which pups are spontaneously aborted (P<.0001). In summary, DMF, a widely used solvent structurally related to DMA and NMP, delays LPS-induced preterm birth in a murine model without overt toxic effects. Re-purposing the DMA/DMF/NMP family of small molecules as anti-inflammatory drugs is a promising new approach to delaying or reducing the incidence of inflammation-induced preterm birth and potentially attenuating other inflammatory disorders as well.


Subject(s)
Dimethylformamide , Premature Birth , Acetamides , Animals , Anti-Inflammatory Agents/pharmacology , Dimethylformamide/adverse effects , Disease Models, Animal , Excipients/adverse effects , Female , Humans , Infant, Newborn , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/prevention & control , Interleukin-6 , Interleukin-8 , Lipopolysaccharides/pharmacology , Mice , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Placenta/metabolism , Pregnancy , Premature Birth/chemically induced , Premature Birth/prevention & control , Solvents/adverse effects , Tumor Necrosis Factor-alpha
4.
Fortune J Health Sci ; 5: 499-509, 2022.
Article in English | MEDLINE | ID: mdl-37886658

ABSTRACT

Inflammatory bowel disease (IBD) affects almost 7 million people worldwide and is increasing in incidence. While the precise pathogenesis of IBD remains unknown, the production of inflammatory cytokines and chemokines play a central role. We have previously found that N, N-dimethylacetamide (DMA), a widely used non-toxic drug excipient, suppresses cytokine and chemokine secretion in vitro and prevents inflammation-induced preterm birth in vivo. Using sandwich enzyme-linked immunosorbent assays (ELISAs), we tested whether DMA attenuates cytokine and chemokine secretion from LPS- or TNFα-stimulated human intestinal epithelial cells and human monocytes and HMGB1 release from RAW 264.7 cells. To test our hypothesis that the mechanism of DMA's effects in in vitro and in vivo models of IBD is inhibition of the NF-κB pathway, we used western blotting to track levels of the nuclear factor kappa B (NF-κB) inhibitory molecule I kappa B alpha (IκBα) in THP-1 human monocytes in the absence or presence of DMA. Finally, we induced colitis in C57Bl/6 mice with dextran sodium sulfate (DSS) and then tested whether i.p injections of DMA at 2.1 g/kg/day attenuates clinical and histopathologic signs of colitis. DMA attenuated cytokine and chemokine release from human intestinal epithelial cells and human monocytes and HMGB1 release from RAW 264.7 cells. Importantly, DMA prevented degradation of IκBα in THP-1 cells, thereby suggesting one mechanism for DMA's effects. Finally, we show here, for the first time, that DMA attenuates clinical and histologic features of DSS-induced colitis. Based on these data, DMA should be further explored in preclinical and clinical trials for its potential as novel drug therapy for IBD.

5.
Front Neurosci ; 15: 687157, 2021.
Article in English | MEDLINE | ID: mdl-34349617

ABSTRACT

Alzheimer disease (AD) is a chronic neurodegenerative disease that accounts for 60-70% of dementia and is the sixth leading cause of death in the United States. The pathogenesis of this debilitating disorder is still not completely understood. New insights into the pathogenesis of AD are needed in order to develop novel pharmacologic approaches. In recent years, numerous studies have shown that insulin resistance plays a significant role in the development of AD. Over 80% of patients with AD have type II diabetes (T2DM) or abnormal serum glucose, suggesting that the pathogenic mechanisms of insulin resistance and AD likely overlap. Insulin resistance increases neuroinflammation, which promotes both amyloid ß-protein deposition and aberrant tau phosphorylation. By increasing production of reactive oxygen species, insulin resistance triggers amyloid ß-protein accumulation. Oxidative stress associated with insulin resistance also dysregulates glycogen synthase kinase 3-ß (GSK-3ß), which leads to increased tau phosphorylation. Both insulin and amyloid ß-protein are metabolized by insulin degrading enzyme (IDE). Defects in this enzyme are the basis for a strong association between T2DM and AD. This review highlights multiple pathogenic mechanisms induced by insulin resistance that are implicated in AD. Several pharmacologic approaches to AD associated with insulin resistance are presented.

6.
Int J Biol Sci ; 17(10): 2652-2665, 2021.
Article in English | MEDLINE | ID: mdl-34326700

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are important in managing lymphoid malignancies by targeting B-cell receptor signaling pathways. Entospletinib (GS-9973) is an oral, selective inhibitor of spleen tyrosine kinase (Syk), currently in the phase II clinical trials for the treatment of chronic lymphocytic leukemia. Syk is abundantly present in the cells of hematopoietic lineage that mediates cell proliferation, differentiation, and adhesion. In this current study, we evaluated the efficacy of GS-9973 to overcome multidrug resistance (MDR) due to the overexpression of the ABCG2 transporter in the non-small cell lung cancer (NSCLC) cell line, NCI-H460/MX20. In vitro, 3 µM of GS-9973 reversed the drug resistance of NCI-H460/MX20 cell line to mitoxantrone or doxorubicin. GS-9973, at 3 µM reverses ABCG2-mediated MDR by blocking ABCG2 efflux activity and downregulating ABCG2 expression at the protein level but did not alter the ABCG2 mRNA expression and subcellular localization of the ABCG2 protein compared to drug-resistant cells incubated with the vehicle. GS-9973 produced a moderate concentration-dependent increase in the ATPase activity of ABCG2 (EC50 = 0.42 µM) and molecular docking data indicated that GS-9973 had a high affinity (-10.226 kcal/mol) for the substrate-binding site of ABCG2. Finally, HPLC analysis proved that the intracellular concentration of GS-9973 is not significantly different in both parental and resistant cell lines. In conclusion, our study suggests that in vitro, GS-9973 in combination with certain anticancer drugs, represent a strategy to overcome ABCG2-mediated MDR cancers.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Indazoles/pharmacology , Lung Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Pyrazines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Humans , Lung Neoplasms/metabolism , Mitoxantrone/pharmacology , Molecular Docking Simulation , Syk Kinase/antagonists & inhibitors
7.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33671108

ABSTRACT

The overexpression of ATP-binding cassette transporter, ABCG2, plays an important role in mediating multidrug resistance (MDR) in certain types of cancer cells. ABCG2-mediated MDR can significantly attenuate or abrogate the efficacy of anticancer drugs by increasing their efflux from cancer cells. In this study, we determined the efficacy of the novel benzamide derivative, VKNG-2, to overcome MDR due to the overexpression of the ABCG2 transporter in the colon cancer cell line, S1-M1-80. In vitro, 5 µM of VKNG-2 reversed the resistance of S1-M1-80 cell line to mitoxantrone (70-fold increase in efficacy) or SN-38 (112-fold increase in efficacy). In contrast, in vitro, 5 µM of VKNG-2 did not significantly alter either the expression of ABCG2, AKT, and PI3K p110ß protein or the subcellular localization of the ABCG2 protein compared to colon cancer cells incubated with the vehicle. Molecular docking data indicated that VKNG-2 had a high docking score (-10.2 kcal/mol) for the ABCG2 transporter substrate-drug binding site whereas it had a low affinity on ABCB1 and ABCC1 transporters. Finally, VKNG-2 produced a significant concentration-dependent increase in ATPase activity (EC50 = 2.3 µM). In conclusion, our study suggests that in vitro, VKNG-2 reverses the resistance of S1-M1-80, a cancer cell line resistant to mitoxantrone and SN-38, by inhibiting the efflux function of the ABCG2 transporter.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Benzamides/chemistry , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Irinotecan/pharmacology , Mitoxantrone/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Resistance, Multiple/drug effects , Humans , Topoisomerase I Inhibitors/pharmacology , Tumor Cells, Cultured
8.
Nanomedicine (Lond) ; 14(21): 2835-2851, 2019 11.
Article in English | MEDLINE | ID: mdl-31793846

ABSTRACT

Aim: Previously, we have shown that inhibition of SphK by the SphK inhibitor-II (SKI II) prevents lipopolysaccharide-induced preterm birth in mice. The aim of this study was to develop a vaginal self-nanoemulsifying drug-delivery system (SNEDDS) for SKI II. Materials & methods: A SKI II-loaded SNEDDS was characterized and tested in a murine preterm birth model. Results: The SNEDDS immediately formed a gel and then slowly emulsified to nanoglobules with over 500-fold enhancement of SKI II solubility at vaginal pH. Intravaginal administration of the SKI II SNEDDS significantly decreased lipopolysaccharide-induced preterm birth in mice. Conclusion: A vaginal nanoformulation of SKI II represents a novel, noninvasive approach to prevent preterm birth.


Subject(s)
Emulsions/chemistry , Enzyme Inhibitors/chemistry , Lipopolysaccharides/metabolism , Nanocapsules/chemistry , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Premature Birth/drug therapy , Animals , Biological Availability , Drug Compounding , Drug Liberation , Drug Stability , Emulsions/pharmacology , Enzyme Inhibitors/metabolism , Female , Humans , Hydrogen-Ion Concentration , Male , Mice , Pregnancy , Solubility , Vagina
9.
Mol Pharm ; 16(10): 4405-4415, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31454253

ABSTRACT

Enterohemorrhagic Escherichia coli O157:H7 (EHEC) or Shiga toxin-producing E. coli (STEC) is known to cause sporadic and epidemic gastrointestinal infections with several incidences of outbreaks. Antibiotic-based therapy further worsens the condition by facilitating the release of Shiga toxins (Stx) and lipopolysaccharides (LPS). Hence, there is an urgent need to develop an antibiotic-free, safe, and effective therapeutic intervention for the treatment of EHEC infections. We proposed a novel therapeutic strategy to address this clinical problem-kill, capture, and inhibit. We aimed to formulate and characterize lauroyl arginate ethyl ester (LAE) and Retro-2 loaded self-nano emulsifying drug delivery systems (SNEDDS). Retro-2 is a recently developed novel class of molecule, which can selectively inhibit retrograde transport of Stx. In this paper, we first carried out preformulation studies of Retro-2, followed by the development of SNEDDS forming arginine anchored nanoglobules (AR-NG), characterization of LPS binding to AR-NG, and finally evaluation of activity against EHEC. Retro-2 showed extremely poor solubility at all gastrointestinal pH values, susceptibility to acidic environments, and good permeability. The positively charged AR-NG spontaneously formed a globule size of 102.8 ± 1.9 nm with a surface charge of +52.15 ± 3 mV and increased the solubility of Retro-2. Further, binding and aggregation of LPS and AR-NG were confirmed by particle size, polydispersity index, zeta potential, fluorescent intensity, turbidity analysis, and a limulus amebocyte lysate (LAL) test. Additionally, a significant reduction in LPS induced TNF-α was observed in AR-NG treated macrophages. Thus, in this paper, we demonstrate a very promising and innovative therapeutic approach based on the "kill (E. Coli), capture (released LPS), and inhibit (transport of Stx)" concept.


Subject(s)
Arginine/chemistry , Benzamides/pharmacology , Disease Outbreaks/prevention & control , Drug Delivery Systems , Enterohemorrhagic Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Nanoparticles/administration & dosage , Thiophenes/pharmacology , Animals , Benzamides/chemistry , Biological Transport , Caco-2 Cells , Cells, Cultured , Colonic Neoplasms/drug therapy , Enterohemorrhagic Escherichia coli/isolation & purification , Escherichia coli Infections/microbiology , Humans , Macrophages/drug effects , Mice , Nanoparticles/chemistry , Thiophenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...