Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Radiat Res ; 183(4): 465-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25807318

ABSTRACT

We have previously demonstrated that the small molecule octadecenyl thiophosphate (OTP), a synthetic mimic of the growth factor-like mediator lysophosphatidic acid (LPA), showed radioprotective activity in a mouse model of total-body irradiation (TBI) when given orally or intraperitoneally 30 min before exposure to 9 Gy γ radiation. In the current study, we evaluated the effects of OTP, delivered subcutaneously, for radioprotection or radiomitigation from -24 h before to up to +72 h postirradiation using a mouse TBI model with therapeutic doses at around 1 mg/kg. OTP was injected at 10 mg/kg without observable toxic side effects in mice, providing a comfortable safety margin. Treatment of C57BL/6 mice with a single dose of OTP over the time period from -12 h before to +26 h after a lethal dose of TBI reduced mortality by 50%. When administered at +48 h to +72 h postirradiation (LD50/30 to LD100/30), OTP reduced mortality by ≥34%. OTP administered at +24 h postirradiation significantly elevated peripheral white blood cell and platelet counts, increased crypt survival in the jejunum, enhanced intestinal glucose absorption and reduced endotoxin seepage into the blood. In the 6.4-8.6 Gy TBI range using LD50/10 as the end point, OTP yielded a dose modification factor of 1.2. The current data indicate that OTP is a potent radioprotector and radiomitigator ameliorating the mortality and tissue injury of acute hematopoietic as well as acute gastrointestinal radiation syndrome.


Subject(s)
Acute Radiation Syndrome/prevention & control , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/radiation effects , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Lysophospholipids/metabolism , Organophosphorus Compounds/pharmacology , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD34/metabolism , Biological Transport/drug effects , Biological Transport/radiation effects , Biomimetic Materials/adverse effects , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/radiation effects , Dose-Response Relationship, Drug , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Glucose/metabolism , HEK293 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Humans , LIM Domain Proteins/metabolism , Leukocyte Count , Mice , Mice, Inbred C57BL , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/pharmacokinetics , Phosphoproteins/metabolism , Platelet Count , Proteasome Endopeptidase Complex , Radiation-Protective Agents/adverse effects , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/pharmacology , Sodium-Hydrogen Exchangers/metabolism , Transcription Factors/metabolism , Whole-Body Irradiation/adverse effects
2.
Biochim Biophys Acta ; 1831(1): 117-25, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127512

ABSTRACT

Due to its antiapoptotic action, derivatives of the lipid mediator lysophosphatidic acid (LPA) provide potential therapeutic utility in diseases associated with programmed cell death. Apoptosis is one of the major pathophysiological processes elicited by radiation injury to the organism. Consequently, therapeutic explorations applying compounds that mimic the antiapoptotic action of LPA have begun. Here we present a brief account of our decade-long drug discovery effort aimed at developing LPA mimics with a special focus on specific agonists of the LPA(2) receptor subtype, which was found to be highly effective in protecting cells from apoptosis. We describe new evidence that 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), a prototypic nonlipid agonist specific to the LPA(2) receptor subtype, rescues apoptotically condemned cells in vitro and in vivo from injury caused by high-dose γ-irradiation. GRI977143 shows the features of a radiomitigator because it is effective in rescuing the lives of mice from deadly levels of radiation when administered 24h after radiation exposure. Our findings suggest that by specifically activating LPA(2) receptors GRI977143 activates the ERK1/2 prosurvival pathway, effectively reduces Bax translocation to the mitochondrion, attenuates the activation of initiator and effector caspases, reduces DNA fragmentation, and inhibits PARP-1 cleavage associated with γ-irradiation-induced apoptosis. GRI977143 also inhibits bystander apoptosis elicited by soluble proapoptotic mediators produced by irradiated cells. Thus, GRI977143 can serve as a prototype scaffold for lead optimization paving the way to more potent analogs amenable for therapeutic exploration. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Subject(s)
Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Receptors, Lysophosphatidic Acid/metabolism , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/pathology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bystander Effect/drug effects , Bystander Effect/radiation effects , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Cytoprotection/drug effects , Cytoprotection/radiation effects , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/radiation effects , Gamma Rays , Lysophospholipids/chemistry , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Knockout , Organophosphorus Compounds/pharmacology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Injuries/pathology , Survival Analysis
3.
Invest Ophthalmol Vis Sci ; 53(4): 2439-45, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22427570

ABSTRACT

Purpose. Super-selective intra-ophthalmic artery chemotherapy (SSIOAC) is an eye-targeted drug-delivery strategy to treat retinoblastoma, the most prevalent primary ocular malignancy in children. Unfortunately, recent clinical reports associate adverse vascular toxicities with SSIOAC using melphalan, the most commonly used chemotherapeutic. Methods. To explore reasons for the unexpected vascular toxicities, we examined the effects of melphalan, as well as carboplatin (another chemotherapeutic used with retinoblastoma), in vitro using primary human retinal endothelial cells, and in vivo using a non-human primate model, which allowed us to monitor the retina in real time during SSIOAC. Results. Both melphalan and carboplatin triggered human retinal endothelial cell migration, proliferation, apoptosis, and increased expression of adhesion proteins intracellullar adhesion molecule-1 [ICAM-1] and soluble chemotactic factors (IL-8). Melphalan increased monocytic adhesion to human retinal endothelial cells. Consistent with these in vitro findings, histopathology showed vessel wall endothelial cell changes, leukostasis, and vessel occlusion. Conclusions. These results reflect a direct interaction of chemotherapeutic drugs with both the vascular endothelium and monocytes. The vascular toxicity may be related to the pH, the pulsatile delivery, or the chemotherapeutic drugs used. Our long-term goal is to determine if changes in the drug of choice and/or delivery procedures will decrease vascular toxicity and lead to better eye-targeted treatment strategies.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Endothelial Cells/drug effects , Melphalan/toxicity , Ophthalmic Artery/drug effects , Animals , Antineoplastic Agents/toxicity , Carboplatin/toxicity , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Macaca mulatta , Neutrophils/metabolism , RNA, Messenger/metabolism
4.
Xenobiotica ; 41(11): 1006-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21864202

ABSTRACT

KZ-41, a quinic acid derivative, significantly reduces mortality in a murine model of hematopoietic acute radiation syndrome. The purpose of this study was to evaluate the systemic pharmacokinetics, elimination, and oral bioavailability of KZ-41 in rats. Male Sprague-Dawley rats (n = 6 per group) received a single dose (10 mg/kg) of KZ-41 administered either intravenously via the jugular vein or orally via gavage. In vitro stability was determined using both rat liver microsomes and the bacteria Gluconobacter oxydans. KZ-41 concentrations were determined using LC-MS/MS (liquid chromatography tandom mass spectrometry). Half-life of KZ-41 was ≈3 hr after either intravenous or oral administration. Mean volume of distribution was 3.3 L/kg. Extent of absorption (F) after oral administration was estimated to be ~100%, which was consistent with the finding that KZ-41 was stable to liver microsomal and bacterial degradation. Following intravenous administration, KZ-41 demonstrated a medium clearance and volume of distribution with a terminal half-life of ≈3 hr. KZ-41 was rapidly and completely absorbed (F ≅ 1), which was consistent with the findings that KZ-41 is resistant to presystemic elimination mechanisms (i.e. enteric bacterial degradation and hepatic metabolism). Thus, KZ-41 represents an excellent candidate for further development as an orally available agent for the mitigation of radiation injury.


Subject(s)
Quinic Acid/analogs & derivatives , Quinic Acid/pharmacokinetics , Radiation-Protective Agents/pharmacokinetics , Administration, Oral , Animals , Chromatography, Liquid , Drug Stability , Injections, Intravenous , Male , Mass Spectrometry , Quinic Acid/blood , Quinic Acid/chemistry , Quinic Acid/pharmacology , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results , Time Factors
5.
Bioorg Med Chem Lett ; 19(18): 5458-60, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19674895

ABSTRACT

Quinic acid (QA) esters found in hot water extracts of Uncaria tomentosa (a.k.a. cat's claw) exert anti-inflammatory activity through mechanisms involving inhibition of the pro-inflammatory transcription factor nuclear factor kappa B (NF-kappaB). Herein, we describe the synthesis and biological testing of novel QA derivatives. Inhibition of NF-kappaB was assessed using A549 (Type II alveolar epithelial-like) cells that stably express a secreted alkaline phosphatase (SEAP) reporter driven by an NF-kappaB response element. A549-NF-kappaB cells were stimulated with TNF-alpha (10 ng/mL) in the presence or absence of QA derivative for 18 hours followed by measurement of SEAP activity. Amide substitution at the carboxylic acid position yielded potent inhibitors of NF-kappaB. A variety of modifications to the amide substitution were tolerated with the N-propyl amide derivative being the most potent. Further examination of the SAR demonstrated that acetylation of the hydroxyl groups reduced NF-kappaB inhibitory activity. QA amide derivatives lacked anti-oxidant activity and were found to be neither anti-proliferative nor cytotoxic at concentrations up to 100 microM. In conclusion, we have discovered a novel series of non-toxic QA amides that potently inhibit NF-kappaB, despite their lack of anti-oxidant activity. Mechanistic studies and pre-clinical efficacy studies in various inflammatory animal models are on-going.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Quinic Acid/chemistry , Quinic Acid/pharmacology , Alkaline Phosphatase/metabolism , Anti-Inflammatory Agents/chemical synthesis , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Pulmonary Alveoli/cytology , Quinic Acid/chemical synthesis , Uncaria/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...