Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Front Neurosci ; 16: 926629, 2022.
Article in English | MEDLINE | ID: mdl-35873810

ABSTRACT

Retinal pigment epithelial (RPE) cells sustain photoreceptor integrity, and when this function is disrupted, retinal degenerations ensue. Herein, we characterize a new cell line from human RPE that we termed ABC. These cells remarkably recapitulate human eye native cells. Distinctive from other epithelia, RPE cells originate from the neural crest and follow a neural development but are terminally differentiated into "epithelial" type, thus sharing characteristics with their neuronal lineages counterparts. Additionally, they form microvilli, tight junctions, and honeycomb packing and express distinctive markers. In these cells, outer segment phagocytosis, phagolysosome fate, phospholipid metabolism, and lipid mediator release can be studied. ABC cells display higher resistance to oxidative stress and are protected from senescence through mTOR inhibition, making them more stable in culture. The cells are responsive to Neuroprotectin D1 (NPD1), which downregulates inflammasomes and upregulates antioxidant and anti-inflammatory genes. ABC gene expression profile displays close proximity to native RPE lineage, making them a reliable cell system to unravel signaling in uncompensated oxidative stress (UOS) and retinal degenerative disease to define neuroprotection sites.

2.
Eur J Med Chem ; 202: 112600, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32629335

ABSTRACT

Although acetaminophen (ApAP) is one of the most commonly used medicines worldwide, hepatotoxicity is a risk with overdose or in patients with compromised liver function. ApAP overdose is the most common cause of acute fulminant hepatic failure. Oxidation of ApAP to N-acetyl-p-benzoquinone imine (NAPQI) is the mechanism for hepatotoxicity. 1 is a non-hepatotoxic, metabolically unstable lipophilic ApAP analog that is not antipyretic. The newly synthesized 3 is a non-hepatotoxic ApAP analog that is stable, lipophilic, and retains analgesia and antipyresis. Intraperitoneal or po administration of the new chemical entities (NCEs), 3b and 3r, in concentrations equal to a toxic dose of ApAP did not result in the formation of NAPQI. Unlike livers from NCE-treated mice, the livers from ApAP-treated mice demonstrated large amounts of nitrotyrosine, a marker of mitochondrial free radical formation, and loss of hepatic tight junction integrity. Given the widespread use of ApAP, hepatotoxicity risk with overuse, and the ongoing opioid epidemic, these NCEs represent a novel, non-narcotic therapeutic pipeline.


Subject(s)
Acetamides/pharmacology , Analgesics/pharmacology , Antipyretics/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Hyperthermia/drug therapy , Liver/drug effects , Acetamides/chemical synthesis , Acetamides/chemistry , Acetic Acid , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Antipyretics/chemical synthesis , Antipyretics/chemistry , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Liver/pathology , Male , Mice , Molecular Structure , Rats , Structure-Activity Relationship
3.
Sci Adv ; 3(9): e1700735, 2017 09.
Article in English | MEDLINE | ID: mdl-28959727

ABSTRACT

We report the characterization of a novel class of lipid mediators termed elovanoids (ELVs) (ELV-N32 and ELV-N34), which are dihydroxylated derivatives of 32:6n3 and 34:6n3, respectively. The precursors of ELVs are made by elongation of a 22:6n3 fatty acid and catalyzed by ELOVL4 (elongation of very-long-chain fatty acids-4). The structure and stereochemistry of ELVs were established using synthetic compounds produced by stereocontrolled total synthesis. We report that ELV-mediated protection is induced in neuronal cultures undergoing either oxygen/glucose deprivation or N-methyl-d-aspartate receptor-mediated excitotoxicity, as well as in experimental ischemic stroke. The methyl ester or sodium salt of ELV-N32 and ELV-N34 resulted in reduced infarct volumes, promoted cell survival, and diminished neurovascular unit disruption when administered 1 hour following 2 hours of ischemia by middle cerebral artery occlusion. Together, our data reveal a novel prohomeostatic and neuroprotective lipid-signaling mechanism aiming to sustain neural cell integrity.


Subject(s)
Homeostasis/drug effects , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Biomarkers , Blood-Brain Barrier/metabolism , Cell Survival/drug effects , Cells, Cultured , Female , Magnetic Resonance Imaging , Molecular Structure , Neuroprotective Agents/chemistry , Pregnancy , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Stereoisomerism , Stroke/drug therapy , Stroke/etiology , Stroke/metabolism , Stroke/physiopathology
4.
Sci Rep ; 7(1): 5279, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28706274

ABSTRACT

Docosahexaenoic acid (DHA, 22:6 n-3) is abundant in the retina and is enzymatically converted into pro-homeostatic docosanoids. The DHA- or eicosapentaenoic acid (EPA)-derived 26 carbon fatty acid is a substrate of elongase ELOVL4, which is expressed in photoreceptor cells and generates very long chain (≥C28) polyunsaturated fatty acids including n-3 (VLC-PUFAs,n-3). While ELOVL4 mutations are linked to vision loss and neuronal dysfunctions, the roles of VLC-PUFAs remain unknown. Here we report a novel class of lipid mediators biosynthesized in human retinal pigment epithelial (RPE) cells that are oxygenated derivatives of VLC-PUFAs,n-3; we termed these mediators elovanoids (ELV). ELVs have structures reminiscent of docosanoids but with different physicochemical properties and alternatively-regulated biosynthetic pathways. The structures, stereochemistry, and bioactivity of ELVs were determined using synthetic materials produced by stereo-controlled chemical synthesis. ELVs enhance expression of pro-survival proteins in cells undergoing uncompensated oxidative stress. Our findings unveil a novel autocrine/paracrine pro-homeostatic RPE cell signaling that aims to sustain photoreceptor cell integrity and reveal potential therapeutic targets for retinal degenerations.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress , Photoreceptor Cells/drug effects , Receptors, Adiponectin/physiology , Retinal Pigment Epithelium/drug effects , Adult , Animals , Cells, Cultured , Eye Proteins/metabolism , Humans , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neuroprotection , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL