Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Neuroreport ; 32(3): 188-197, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33470761

ABSTRACT

Pyroptosis has been reported to contribute to the traumatic brain injury (TBI) process. Ac-FLTD-CMK is a newly synthesized pyroptosis inhibitor. However, whether Ac-FLTD-CMK inhibits pyroptosis and plays a neuroprotective role after TBI is unknown. The present study aimed to determine the effects of Ac-FLTD-CMK on TBI in a mouse model. Male C57BL/6 mice were randomly divided into sham, TBI + vehicle, and TBI + Ac-FLTD-CMK groups. TBI was induced using a weight-drop apparatus. Intraventricular injection of Ac-FLTD-CMK was performed 30 min after TBI. Caspase-1, caspase-11, gasdermin-D (GSDMD), and caspase-3 expression in the peri-contusional cortex were assessed by western blotting. Interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) expression in the peri-contusional cortex were measured using ELISA. Behavioral experiments, brain water content, Evans blue extravasation, lactate dehydrogenase (LDH) release, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining were also performed. The results showed that Ac-FLTD-CMK administration significantly downregulated caspase-1 p20, caspase-11 p20, GSDMD N-terminal, IL-1ß, and IL-18 expression; reduced LDH release; alleviated neuronal death; attenuated brain edema and blood-brain barrier damage; and improved neurobehavioral function. These findings indicate that Ac-FLTD-CMK treatment suppresses pyroptosis and protects mice against TBI.


Subject(s)
Brain Contusion/metabolism , Brain/drug effects , Caspase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Injuries, Traumatic/metabolism , Caspase 1/drug effects , Caspase 1/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspases, Initiator/drug effects , Caspases, Initiator/metabolism , Disease Models, Animal , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Open Field Test , Phosphate-Binding Proteins/drug effects , Phosphate-Binding Proteins/metabolism , Rotarod Performance Test
2.
Alcohol Clin Exp Res ; 44(8): 1585-1597, 2020 08.
Article in English | MEDLINE | ID: mdl-32524615

ABSTRACT

BACKGROUND: Alcohol use disorders affect millions of people worldwide, and there is growing evidence that excessive alcohol intake causes severe damage to the brain of both humans and animals. Numerous studies on chronic alcohol exposure in animal models have identified that many functional impairments are associated with the hippocampus, which is a structure exhibiting substantial vulnerability to alcohol exposure. However, the precise mechanisms that lead to structural and functional impairments of the hippocampus are poorly understood. Herein, we report a novel cell death type, namely pyroptosis, which accounts for alcohol neurotoxicity in mice. METHODS: For this study, we used an in vivo model to induce alcohol-related neurotoxicity in the hippocampus. Adult male C57BL/6 mice were treated with 95% alcohol vapor either alone or in combination with selective cannabinoid receptor antagonists or agonists, and VX765 (Belnacasan), which is a selective caspase-1 inhibitor. RESULTS: Alcohol-induced in vivo pyroptosis occurs because of an increase in the levels of pyroptotic proteins such as nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3), caspase-1, gasdermin D (GSDMD), and amplified inflammatory response. Our results indicated that VX765 suppressed the expression of caspase-1 and inhibited the maturation of the proinflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Additionally, chronic alcohol intake created an imbalance in the endocannabinoid system and regulated 2 cannabinoid receptors (CB1R and CB2R) in the hippocampus. Specific antagonists of CB1R (AM251 and AM281) significantly ameliorated alcohol-induced pyroptosis signaling and inactivated the inflammatory response. CONCLUSIONS: Alcohol induces hippocampal pyroptosis, which leads to neurotoxicity, thereby indicating that pyroptosis may be an essential pathway involved in chronic alcohol-induced hippocampal neurotoxicity. Furthermore, cannabinoid receptors are regulated during this process, which suggests promising therapeutic strategies against alcohol-induced neurotoxicity through pharmacologic inhibition of CB1R.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Cannabinoid Receptor Agonists/pharmacology , Caspase 1/drug effects , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Dipeptides/pharmacology , Inflammation , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Morpholines/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurotoxicity Syndromes , Phosphate-Binding Proteins/drug effects , Phosphate-Binding Proteins/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , para-Aminobenzoates/pharmacology
3.
Inflamm Bowel Dis ; 24(9): 1978-1991, 2018 08 16.
Article in English | MEDLINE | ID: mdl-29718324

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) afflicts 5 million people and is increasing in prevalence. There is an unmet clinical need for safer and effective treatments for IBD. The BT-11 is a small molecule oral therapeutic that ameliorates IBD by targeting lanthionine synthetase C-like 2 (LANCL2) and has a benign safety profile in rats. METHODS: Mdr1a-/-, dextran sodium sulphate , and adoptive transfer mouse models of colitis were employed to validate therapeutic efficacy and characterize the mechanisms of therapeutic efficacy of BT-11. In vitro cultures of CD4+ T cell differentiation and human peripheral blood mononuclear cells from Crohn's disease patients were used to determine its potential for human translation. RESULTS: BT-11 reduces inflammation in multiple mouse models of IBD. Oral treatment with BT-11 increases the numbers of lamina propria regulatory T cells (Tregs) in a LANCL2-dependent manner. In vitro, BT-11 increases the differentiation in Treg phenotypes, the upregulation of genes implicated in Treg cell stability, and conditions Treg cells to elicit greater suppressive actions. These immunoregulatory effects are intertwined with the ability of BT-11 to regulate late stage glycolysis and tricarboxylic acid cycle. Immunometabolic mechanistic findings translate into human peripheral blood mononuclear cells from healthy individuals and Crohn's disease patients. CONCLUSIONS: BT-11 is a safe, efficacious oral therapeutic for IBD with a human translatable mechanism of action that involves activation of LANCL2, immunometabolic modulation of CD4+ T cell subsets leading to stable regulatory phenotypes in the colonic LP.


Subject(s)
Benzimidazoles/administration & dosage , Colitis/drug therapy , Colitis/immunology , Membrane Proteins/drug effects , Phosphate-Binding Proteins/drug effects , Piperazines/administration & dosage , T-Lymphocytes, Regulatory/drug effects , Adoptive Transfer , Animals , Cell Culture Techniques , Colitis/chemically induced , Colon/drug effects , Crohn Disease/drug therapy , Crohn Disease/immunology , Dextran Sulfate , Disease Models, Animal , Humans , Intestinal Mucosa/drug effects , Mice , Phenotype , T-Lymphocyte Subsets/drug effects
4.
Semin Dial ; 19(3): 195-9, 2006.
Article in English | MEDLINE | ID: mdl-16689967

ABSTRACT

Accumulation of inorganic phosphate due to renal functional impairment contributes to the increased cardiovascular mortality observed in dialysis patients. Phosphate plays a causative role in the development of vascular calcification in renal failure; treatment with calcium-based phosphate binders and vitamin D can further increase the Ca x PO(4) product and add to the risk of ectopic mineralization. The new generation of calcium-free phosphate binders, sevelamer and lanthanum, can control hyperphosphatemia without adding to the patients calcium load. In this article, the metabolism of lanthanum carbonate and its effects in bone, liver and brain are discussed. Although lanthanum is a metal cation its effects are not comparable to those of aluminum. Indeed, in clinical studies no toxic effects of lanthanum have been reported after up to four years of follow-up. The bioavailability of lanthanum is extremely low. The effects observed in bone are due to phosphate depletion, with no signs of direct bone toxicity yet observed in rats or humans. The liver is the main route of excretion for lanthanum carbonate, which can be localized in the lysosomes of hepatocytes. No lanthanum could be detected in brain tissue.


Subject(s)
Lanthanum/metabolism , Phosphate-Binding Proteins/drug effects , Animals , Biological Availability , Bone and Bones/drug effects , Bone and Bones/metabolism , Brain/drug effects , Brain/metabolism , Calcinosis/drug therapy , Calcinosis/metabolism , Clinical Trials as Topic , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/metabolism , Lanthanum/pharmacokinetics , Liver/drug effects , Liver/metabolism , Phosphate-Binding Proteins/blood , Phosphate-Binding Proteins/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...