Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cells ; 12(11)2023 06 05.
Article in English | MEDLINE | ID: mdl-37296668

ABSTRACT

BACKGROUND: Cardiac arrest (CA) can lead to neuronal degeneration and death through various pathways, including oxidative, inflammatory, and metabolic stress. However, current neuroprotective drug therapies will typically target only one of these pathways, and most single drug attempts to correct the multiple dysregulated metabolic pathways elicited following cardiac arrest have failed to demonstrate clear benefit. Many scientists have opined on the need for novel, multidimensional approaches to the multiple metabolic disturbances after cardiac arrest. In the current study, we have developed a therapeutic cocktail that includes ten drugs capable of targeting multiple pathways of ischemia-reperfusion injury after CA. We then evaluated its effectiveness in improving neurologically favorable survival through a randomized, blind, and placebo-controlled study in rats subjected to 12 min of asphyxial CA, a severe injury model. RESULTS: 14 rats were given the cocktail and 14 received the vehicle after resuscitation. At 72 h post-resuscitation, the survival rate was 78.6% among cocktail-treated rats, which was significantly higher than the 28.6% survival rate among vehicle-treated rats (log-rank test; p = 0.006). Moreover, in cocktail-treated rats, neurological deficit scores were also improved. These survival and neurological function data suggest that our multi-drug cocktail may be a potential post-CA therapy that deserves clinical translation. CONCLUSIONS: Our findings demonstrate that, with its ability to target multiple damaging pathways, a multi-drug therapeutic cocktail offers promise both as a conceptual advance and as a specific multi-drug formulation capable of combatting neuronal degeneration and death following cardiac arrest. Clinical implementation of this therapy may improve neurologically favorable survival rates and neurological deficits in patients suffering from cardiac arrest.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest , Animals , Rats , Cardiopulmonary Resuscitation/methods , Heart Arrest/complications , Heart Arrest/therapy , Rats, Sprague-Dawley , Rodentia
2.
Nat Commun ; 13(1): 7127, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36443291

ABSTRACT

Peptides, polymers of amino acids, comprise a vital and expanding therapeutic approach. Their rapid degradation by proteases, however, represents a major limitation to their therapeutic utility and chemical modifications to native peptides have been employed to mitigate this weakness. Herein, we describe functionalized thiocarbazate scaffolds as precursors of aza-amino acids, that, upon activation, can be integrated in a peptide sequence to generate azapeptides using conventional peptide synthetic methods. This methodology facilitates peptide editing-replacing targeted amino acid(s) with aza-amino acid(s) within a peptide-to form azapeptides with preferred therapeutic characteristics (extending half-life/bioavailability, while at the same time typically preserving structural features and biological activities). We demonstrate the convenience of this azapeptide synthesis platform in two well-studied peptides with short half-lives: FSSE/P5779, a tetrapeptide inhibitor of HMGB1/MD-2/TLR4 complex formation, and bradykinin, a nine-residue vasoactive peptide. This bench-stable thiocarbazate platform offers a robust and universal approach to optimize peptide-based therapeutics.


Subject(s)
Amino Acids , Bradykinin , Half-Life , Peptide Hydrolases , Endopeptidases
3.
Curr Med Chem ; 29(42): 6336-6358, 2022.
Article in English | MEDLINE | ID: mdl-35538801

ABSTRACT

For over 50 years of azapeptide synthetic techniques, developments have renewed the field of peptidomimetic therapeutics. Azapeptides are close surrogates of natural peptides: they contain a substitution of the amino acid α-carbon by a nitrogen atom. Goserelin (1989) and Atazanavir (2003) are two well-known, FDA-approved azapeptide-based drugs for the treatment of cancers and HIV infection, providing evidence for the successful clinical implementation of this class of therapeutic. This review highlights the azapeptides in recent medicinal chemistry applications and synthetic milestones. We describe the current techniques for azapeptide bond formation by introducing azapeptide coupling reagents and chain elongation methods both in solution and solid-phase strategies.


Subject(s)
Aza Compounds , HIV Infections , Peptidomimetics , Humans , Aza Compounds/chemistry , Atazanavir Sulfate , Goserelin , Peptides/chemistry , Amino Acids/chemistry , Carbon , Nitrogen
4.
Mol Med ; 26(1): 103, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33167852

ABSTRACT

The response to viral infection generally includes an activation of the adaptive immune response to produce cytotoxic T cells and neutralizing antibodies. We propose that SARS-CoV-2 activates the innate immune system through the renin-angiotensin and kallikrein-bradykinin pathways, blocks interferon production and reduces an effective adaptive immune response. This model has therapeutic implications.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Immunity, Innate , Pneumonia, Viral/immunology , Animals , Bradykinin/metabolism , COVID-19 , Humans , Kallikreins/metabolism , Models, Immunological , Pandemics , Renin-Angiotensin System , SARS-CoV-2
5.
Future Med Chem ; 12(18): 1647-1656, 2020 09.
Article in English | MEDLINE | ID: mdl-32672061

ABSTRACT

During a disease outbreak/pandemic situation such as COVID-19, researchers are in a prime position to identify and develop peptide-based therapies, which could be more rapidly and cost-effectively advanced into a clinical setting. One drawback of natural peptide drugs, however, is their proteolytic instability; peptidomimetics can help to overcome this caveat. In this review, we summarize peptide and peptide-based therapeutics that target one main entry pathway of SARS-CoV-2, which involves the host ACE2 receptor and viral spike (S) protein interaction. Furthermore, we discuss the advantages of peptidomimetics and other potential targets that have been studied using peptide-based therapeutics for COVID-19.


Subject(s)
Antiviral Agents/therapeutic use , Coronavirus Infections/drug therapy , Peptides/therapeutic use , Peptidomimetics/therapeutic use , Pneumonia, Viral/drug therapy , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Humans , Pandemics , Peptidyl-Dipeptidase A/drug effects , Spike Glycoprotein, Coronavirus/drug effects , Virus Internalization
6.
Trends Immunol ; 40(5): 447-462, 2019 05.
Article in English | MEDLINE | ID: mdl-30962001

ABSTRACT

Produced by many cell types, macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with critical and supporting roles in many disease states and conditions. Its disease associations, myriad functions, receptors, and downstream signaling have been the subject of considerable research, yet many questions remain. Moreover, the relevance of MIF's partially functionally redundant family member, D-dopachrome tautomerase (D-DT), also remains to be further characterized. Here, we discuss recent discoveries demonstrating direct roles of MIF in supporting NLR Family Pyrin Domain-Containing 3 (NRLP3) inflammasome activation, as well as acting as a molecular chaperone for intracellular proteins. These findings may offer new clues to understanding MIF's multiple functions, and assist the development of putative MIF-targeting therapeutics for a variety of pathologies.


Subject(s)
Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Humans , Inflammasomes/immunology , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
7.
J Biomol Struct Dyn ; 37(14): 3721-3730, 2019 09.
Article in English | MEDLINE | ID: mdl-30238832

ABSTRACT

Supplemental data for this article can be accessed here.High mobility group box-1 (HMGb1), an endogenous danger-associated molecular pattern protein (DAMP) whose extracellular release has been associated with sterile injury and various inflammatory diseases and conditions, has been shown to be a valuable clinical drug target. Elucidation of the specific interactions with the HMGb1 receptor, Toll-like receptor 4 (TLR4) and adaptor protein myeloid differentiation factor-2 (MD-2), will lead to more precisely targeted therapeutics. We sought to examine detailed interactions and dynamics of the HMGb1 A-box and B-box fragments, as well as the intact protein using in silico protein-protein docking (ZDOCK, ZRANK) and molecular dynamics (Schrödinger Desmond, New York, NY). Mutagenesis and SPR-binding studies allowed us to draw further conclusions regarding the details of the HMGb1-TLR4-MD2 interaction and shed light on the reasons for the opposing biological activities of HMGb1 A-box and B-box fragments. From our findings, we hypothesize that disulfide A-box fragment binds as an anchor toward the TLR4-MD-2 but does not facilitate the TLR4 dimer formation, thereby competing with the HMGb1-binding site and preventing HMGb1-induced signaling and downstream inflammation, whereas the pro-inflammatory B-box fragment retains the MD-2 active conformation and binds to both TLR4 proteins in the complex to aid TLR4 dimer formation, which activates the intracellular signaling for downstream inflammatory pathways and cytokine release. Communicated by Ramaswamy H. Sarma.


Subject(s)
HMGB1 Protein/metabolism , Protein Multimerization , Toll-Like Receptor 4/metabolism , Animals , Disulfides/metabolism , HMGB1 Protein/chemistry , Humans , Lymphocyte Antigen 96/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Rats , Toll-Like Receptor 4/chemistry
8.
J Med Chem ; 61(12): 5093-5107, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29268019

ABSTRACT

High mobility group box-1 (HMGb1) protein, a nuclear non-histone protein that is released or secreted from the cell in response to damage or stress, is a sentinel for the immune system that plays a critical role in cell survival/death pathways. This review highlights key features of the endogenous danger-associated molecular pattern (DAMP) protein, HMGb1 in the innate inflammatory response along with various cofactors and receptors that regulate its downstream effects. The evidence demonstrating increased levels of HMGb1 in human inflammatory diseases and conditions is presented, along with a summary of current small molecule or peptide-like antagonists proven to specifically target HMGb1. Additionally, we delineate the measures needed toward validating this protein as a clinically relevant biomarker or bioindicator and as a relevant drug target.


Subject(s)
HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , Inflammation/immunology , Molecular Targeted Therapy/methods , Alarmins/immunology , Alarmins/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Humans , Immunity, Innate/drug effects , Immunity, Innate/physiology , Inflammation/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism
9.
Bioorg Med Chem Lett ; 27(20): 4725-4729, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28927788

ABSTRACT

FISLE-412 is the first reported small molecule peptidomimetic that neutralizes anti-dsDNA autoantibodies associated with systemic lupus erythematosus (SLE) pathogenesis. FISLE-412 is a complex small molecule that involves a challenging synthesis scheme, but has attractive pharmacological activities as a potential small molecule therapeutic in lupus. Therefore, we initiated a Structure-Activity Relationship study to simplify the complexity of FISLE-412. We synthesized a small library of mimetopes around the FISLE-412 structure and identified several analogues which could neutralize anti-DNA lupus antibodies in vitro and ex vivo. Our strategies reduced the structural complexity of FISLE-412 and provide important information that may guide development of potential autoantibody-targeted lupus therapeutics.


Subject(s)
Antibodies, Antinuclear/metabolism , Quinolines/chemistry , Saquinavir/chemistry , Antibodies, Antinuclear/chemistry , Drug Design , Humans , Inhibitory Concentration 50 , Kidney/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Peptidomimetics , Polyamines/chemistry , Polyamines/metabolism , Quinolines/metabolism , Structure-Activity Relationship
10.
Drug Discov Today ; 22(1): 97-104, 2017 01.
Article in English | MEDLINE | ID: mdl-27666193

ABSTRACT

Acetylcholine, a major neurotransmitter of the parasympathetic and sympathetic nervous systems, was discovered in the early 1900s. Over the years, researchers have revealed much about its regulation, properties of its receptors and features of the downstream signaling that influence its terminal effects. The acetylcholine system, traditionally associated with neuromuscular communication, is now known to play a crucial part in modulation of the immune system and other 'rest and digest' effects. Recent research seeks to elucidate the system's role in brain functions including cognition, sleep, arousal, motivation, reward and pain. We highlight clinically approved and experimental drugs that modulate the acetylcholine receptors. The complexities in targeting the acetylcholine receptors are vast and finding future indications for drug development associated with specific acetylcholine receptors remains a challenge.


Subject(s)
Acetylcholine/metabolism , Cholinergic Agonists/pharmacology , Cholinergic Antagonists/pharmacology , Drug Discovery/methods , Receptors, Cholinergic/metabolism , Acetylcholine/genetics , Animals , Cholinergic Agonists/therapeutic use , Cholinergic Antagonists/therapeutic use , Humans , Ligands , Molecular Targeted Therapy , Receptors, Cholinergic/genetics , Signal Transduction
11.
J Med Chem ; 59(19): 8859-8867, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27603688

ABSTRACT

Systemic lupus erythematosus is an autoimmune disease that can affect numerous tissues and is characterized by the production of nuclear antigen-directed autoantibodies (e.g., anti-dsDNA). Using a combination of virtual and ELISA-based screens, we made the intriguing discovery that several HIV-protease inhibitors can function as decoy antigens to specifically inhibit the binding of anti-dsDNA antibodies to target antigens such as dsDNA and pentapeptide DWEYS. Computational modeling revealed that HIV-protease inhibitors comprised structural features present in DWEYS and predicted that analogues containing more flexible backbones would possess preferred binding characteristics. To address this, we reduced the internal amide backbone to improve flexibility, producing new small-molecule decoy antigens, which neutralize anti-dsDNA antibodies in vitro, in situ, and in vivo. Pharmacokinetic and SLE model studies demonstrated that peptidomimetic FISLE-412,1 a reduced HIV protease inhibitor analogue, was well-tolerated, altered serum reactivity to DWEYS, reduced glomeruli IgG deposition, preserved kidney histology, and delayed SLE onset in NZB/W F1 mice.


Subject(s)
Antibodies, Antinuclear/immunology , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Animals , DNA/immunology , Drug Discovery , Female , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/pharmacology , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred NZB , Models, Molecular
12.
Physiol Rep ; 4(24)2016 12.
Article in English | MEDLINE | ID: mdl-28039410

ABSTRACT

Pulmonary hypertension (PH) is a complex disease comprising a pathologic remodeling and thickening of the pulmonary vessels causing an after load on the right heart ventricle that can result in ventricular failure. Triggered by oxidative stress, episodes of hypoxia, and other undetermined causes, PH is associated with poor outcomes and a high rate of morbidity. In the neonate, this disease has a similar etiology but is further complicated by the transition to breathing after birth, which requires a reduction in vascular resistance. Persistent pulmonary hypertension of the newborn (PPHN) is one form of PH that is frequently unresponsive to current therapies including inhaled nitric oxide (due to lack of proper absorption and diffusion), and other therapeutics targeting signaling mediators in vascular endothelium and smooth muscle. The need for novel agents, which target distinct pathways in pulmonary hypertension, remains. Herein, we investigated the therapeutic effects of novel muscarinic receptor ligand C1213 in models of PH We demonstrated that via M3 muscarinic receptors, C1213 induced activating- eNOS phosphorylation (serine-1177), which is known to lead to nitric oxide (NO) production in endothelial cells. Using signaling pathway inhibitors, we discovered that AKT and calcium signaling contributed to eNOS phosphorylation induced by C1213. As expected for an eNOS-stimulating agent, in ex vivo and in vivo models, C1213 triggered pulmonary vasodilation and induced both pulmonary artery and systemic blood pressure reductions demonstrating its potential value in PH and PPHN In brief, this proof-of-concept study provides evidence that an M3 muscarinic receptor functionally selective ligand stimulates downstream pathways leading to antihypertensive effects using in vitro, ex vivo, and in vivo models of PH.


Subject(s)
Antihypertensive Agents/pharmacology , Benzylamines/pharmacology , Endothelial Cells/drug effects , Guanidines/pharmacology , Hypertension, Pulmonary/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptor, Muscarinic M3/metabolism , Animals , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Cell Hypoxia/drug effects , Cell Line , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Hypertension, Pulmonary/prevention & control , Oncogene Protein v-akt/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/antagonists & inhibitors , Sheep , Signal Transduction/drug effects
13.
Eur J Pharmacol ; 740: 683-9, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-24967533

ABSTRACT

Macrophage migration inhibitory factor is a multifunctional cytokine involved in the regulation of immune processes and also in apoptosis induction. Elevated MIF expression is detrimental for insulin-producing beta cells and MIF inhibition protected beta cells from several cytotoxic insults such as inflammatory cytokines, high fatty acids or high glucose concentrations. Therefore, the aim of this study was to investigate two newly synthesized small molecule MIF inhibitors (K664-1 and K647-1) and to compare them with previously established effects of the prototypical MIF inhibitor, ISO-1. Our results indicate that K664-1 and K647-1 are 160- and 40-fold more effective in inhibition of MIF׳s tautomerase activity than ISO-1. Also, new inhibitors confer beta cell protection from cytokine-triggered apoptosis at significantly lower concentrations than ISO-1. Although all three MIF inhibitors inhibit caspase 3 activity, K664-1 and K647-1 suppress pro-apoptotic BAX protein expression and up-regulate anti-apoptotic Bcl-2 mRNA. Finally, all three MIF inhibitors operate through blockade of nitric oxide production stimulated by cytokines. In conclusion, two novel MIF inhibitors are more potent than ISO-1 and operate through inhibition of the mitochondria-related apoptotic pathway. We propose that these compounds represent a unique class of anti-MIF antagonists that should be further tested for therapeutic use.


Subject(s)
Catechols/pharmacology , Heterocyclic Compounds, 2-Ring/pharmacology , Insulin-Secreting Cells/drug effects , Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Niacin/analogs & derivatives , Oximes/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Intramolecular Oxidoreductases/genetics , Isoxazoles/pharmacology , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Niacin/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Rats , Tumor Necrosis Factor-alpha/pharmacology
14.
Arch Pharm (Weinheim) ; 347(2): 104-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24243226

ABSTRACT

Macrophage migration inhibitory factor (MIF), a cytokine involved in the pathogenesis of sepsis, diabetes, asthma, arthritis, and cancer, has an enzymatic active site that has proven to be an effective target for small molecule-based inhibitors. Herein, we describe the synthesis of a novel arylazoarylmethane compound (3) from a known arylhydrazone MIF inhibitor (2). This new compound has improved stability and in vivo biological activity.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Hydrazones/chemical synthesis , Hydrazones/pharmacology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Animals , Carrageenan , Disease Models, Animal , Drug Design , Drug Stability , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/prevention & control , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Molecular Structure , Structure-Activity Relationship
15.
J Mol Graph Model ; 42: 104-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23603608

ABSTRACT

Recent research suggested the involvement of migration inhibitor factor (MIF) in cancer and inflammatory diseases, which prompted several attempts to develop new MIF inhibitors. Accordingly, we investigated the pharmacophoric space of 79 MIF inhibitors using seven diverse subsets of inhibitors to identify plausible binding hypotheses (pharmacophores). Subsequently, we implemented genetic algorithm and multiple linear regression analysis to select optimal combination of pharmacophores and physicochemical descriptors capable of explaining bioactivity variation within the training compounds (QSAR model, r63=0.62, F=42.8, rLOO(2)=0.721,rPRESS(2) against 16 external test inhibitors=0.58). Two orthogonal pharmacophores appeared in the optimal QSAR model suggestive of at least two binding modes available to ligands inside MIF binding pocket. Subsequent validation using receiver operating characteristic (ROC) curves analysis established the validity of these two pharmacophores. We employed these pharmacophoric models and associated QSAR equation to screen the National Cancer Institute (NCI) list of compounds. Eight compounds gave >50% inhibition at 100µM. Two molecules illustrated >75% inhibition at 10µM.


Subject(s)
Drug Design , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/chemistry , Quantitative Structure-Activity Relationship , Binding Sites , Computer Simulation , Humans , Inflammation/drug therapy , Models, Molecular , Neoplasms/drug therapy , Protein Binding , ROC Curve
16.
Proc Natl Acad Sci U S A ; 108(20): 8224-7, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21536912

ABSTRACT

Abnormally low plasma concentrations of thyroid hormones during sepsis often occur in the absence of thyroidal illness; however, the mechanisms involved in the "euthyroid sick syndrome" remain poorly understood. Here, we describe a previously unrecognized interaction between the thyroid hormone thyroxine (T(4)) and the proinflammatory cytokine macrophage migration inhibitory factor (MIF), together with its clinical relevance in sepsis. We found that in both patients with severe sepsis, and our rodent model, low plasma T(4) concentrations were inversely correlated with plasma MIF concentrations. The MIF molecule contains a hydrophobic pocket that is important for many of its proinflammatory activities. Binding of L-T(4) (or its hormonally inert isomer D-T(4)) significantly, and dose-dependently, inhibited the catalytic activity of this pocket. Moreover, administration of exogenous D-T(4) significantly improved survival in mice with severe sepsis. To examine the specificity of the MIFT(4) interaction, wild-type and MIF knockout mice were subjected to the carrageenan-air pouch model of inflammation and then treated with D-T(4) or vehicle. D-T(4) significantly inhibited leukocyte infiltration in wild-type mice but not in MIF knockout mice, providing evidence that in vivo T(4) may influence MIF-mediated inflammatory responses via inhibition of its hydrophobic proinflammatory pocket. These findings demonstrate a new physiological role for T(4) as a natural inhibitor of MIF proinflammatory activity. The data may also, in part, explain the low plasma T(4) concentrations in critically ill, euthyroid patients and suggest that targeting the imbalance between MIF and T(4) may be beneficial in improving outcome from sepsis.


Subject(s)
Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Thyroxine/physiology , Animals , Binding Sites , Humans , Inflammation , Macrophage Migration-Inhibitory Factors/blood , Mice , Mice, Knockout , Sepsis/blood , Sepsis/drug therapy , Survival Rate , Thyroid Hormones/blood , Thyroxine/blood , Thyroxine/therapeutic use
17.
Future Med Chem ; 3(1): 45-63, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21428825

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pleiotropic proinflammatory cytokine that has been implicated as playing a causative role in many disease states, including sepsis, pneumonia, diabetes, rheumatoid arthritis, inflammatory bowel disease, psoriasis and cancer. To inhibit the enzymatic and biologic activities of MIF, we and others have developed small-molecule MIF inhibitors. Most MIF inhibitors bind within the hydrophobic pocket that contains highly conserved amino acids known to be essential for MIF's proinflammatory activity. The best characterized of these small-molecule MIF inhibitors, (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) has been validated in scores of laboratories worldwide. Like neutralizing anti-MIF antibodies, ISO-1 significantly improves survival and reduces disease progression and/or severity in multiple murine models where MIF is implicated. This MIF inhibitor, its derivatives and other MIF-targeted compounds show great promise for future testing in disease states where increased MIF activity has been discovered.


Subject(s)
Drug Discovery/methods , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/immunology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Disease Models, Animal , Humans , Macrophage Migration-Inhibitory Factors/chemistry , Models, Molecular
18.
Biochem J ; 379(Pt 2): 229-33, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-14984364

ABSTRACT

To elucidate the control of hepatic cholesterol metabolism by leptin, rats were administered IV (intravenous) leptin, ICV (intracerebroventricular) leptin or saline. A single low dose of ICV leptin was as effective as a continuous IV infusion of high-dose leptin at decreasing the activities of 3-hydroxy-3-methylglutaryl-CoA reductase and cholesterol 7alpha-hydroxylase. These results indicate that the hepatic response to leptin is transduced via the central nervous system.


Subject(s)
Cholesterol/metabolism , Leptin/pharmacology , Liver/metabolism , Animals , Cerebral Ventricles , Cholesterol/blood , Cholesterol 7-alpha-Hydroxylase/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Infusions, Intravenous , Infusions, Parenteral , Leptin/administration & dosage , Lipids/blood , Lipoproteins/blood , Liver/drug effects , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...