Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Int J Mol Sci ; 24(11)2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37298571

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is an enzyme crucially implicated in aberrations of various signaling pathways that underlie the development of different human pathologies, such as obesity, diabetes, cancer, and neurodegenerative disorders. Its inhibition can prevent these pathogenetic events, thus providing a useful tool for the discovery of novel therapeutic agents. The search for allosteric PTP1B inhibitors can represent a successful strategy to identify drug-like candidates by offering the opportunity to overcome some issues related to catalytic site-directed inhibitors, which have so far hampered the development of drugs targeting this enzyme. In this context, trodusquemine (MSI-1436), a natural aminosterol that acts as a non-competitive PTP1B inhibitor, appears to be a milestone. Initially discovered as a broad-spectrum antimicrobial agent, trodusquemine exhibited a variety of unexpected properties, ranging from antidiabetic and anti-obesity activities to effects useful to counteract cancer and neurodegeneration, which prompted its evaluation in several preclinical and clinical studies. In this review article, we provide an overview of the main findings regarding the activities and therapeutic potential of trodusquemine and their correlation with PTP1B inhibition. We also included some aminosterol analogues and related structure-activity relationships that could be useful for further studies aimed at the discovery of new allosteric PTP1B inhibitors.


Subject(s)
Neoplasms , Phosphoric Monoester Hydrolases , Humans , Hypoglycemic Agents/pharmacology , Obesity/drug therapy , Obesity/metabolism , Drug Discovery , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Enzyme Inhibitors/pharmacology
2.
Eur J Med Chem ; 252: 115270, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36934484

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a serious chronic disease with an alarmingly growing worldwide prevalence. Current treatment of T2DM mainly relies on drug combinations in order to control blood glucose levels and consequently prevent the onset of hyperglycaemia-related complications. The development of multiple-targeted drugs recently emerged as an attractive alternative to drug combinations for the treatment of complex diseases with multifactorial pathogenesis, such as T2DM. Protein tyrosine phosphatase 1B (PTP1B) and aldose reductase (AKR1B1) are two enzymes crucially involved in the development of T2DM and its chronic complications and, therefore, dual inhibitors targeted to both these enzymes could provide novel agents for the treatment of this complex pathological condition. In continuing our search for dual-targeted PTP1B/AKR1B1 inhibitors, we designed new (5-arylidene-4-oxo-2-thioxothiazolidin-3-yl)alkanoic acids. Among them, 3-(4-phenylbutoxy)benzylidene derivatives 6f and 7f, endowed with interesting inhibitory activity against both targets, proved to control specific cellular pathways implicated in the development of T2DM and related complications.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/drug therapy , Phosphoric Monoester Hydrolases , Ligands , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Aldehyde Reductase
3.
J Med Chem ; 65(16): 10848-10881, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35924548

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT-2) inhibitors (gliflozins) represent the most recently approved class of oral antidiabetic drugs. SGLT-2 overexpression in diabetic patients contributes significantly to hyperglycemia and related complications. Therefore, SGLT-2 became a highly interesting therapeutic target, culminating in the approval for clinical use of dapagliflozin and analogues in the past decade. Gliflozins improve glycemic control through a novel insulin-independent mechanism of action and, moreover, exhibit significant cardiorenal protective effects in both diabetic and nondiabetic subjects. Therefore, gliflozins have received increasing attention, prompting extensive structure-activity relationship studies and optimization approaches. The discovery that intestinal SGLT-1 inhibition can provide a novel opportunity to control hyperglycemia, through a multifactorial mechanism, recently encouraged the design of low adsorbable inhibitors selectively directed to the intestinal SGLT-1 subtype as well as of dual SGLT-1/SGLT-2 inhibitors, representing a compelling strategy to identify new antidiabetic drug candidates.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Sodium-Glucose Transporter 2 Inhibitors , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Humans , Hyperglycemia/complications , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Sodium , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
4.
Molecules ; 26(2)2021 Jan 10.
Article in English | MEDLINE | ID: mdl-33435264

ABSTRACT

Diabetes mellitus (DM) is a complex disease which currently affects more than 460 million people and is one of the leading cause of death worldwide. Its development implies numerous metabolic dysfunctions and the onset of hyperglycaemia-induced chronic complications. Multiple ligands can be rationally designed for the treatment of multifactorial diseases, such as DM, with the precise aim of simultaneously controlling multiple pathogenic mechanisms related to the disease and providing a more effective and safer therapeutic treatment compared to combinations of selective drugs. Starting from our previous findings that highlighted the possibility to target both aldose reductase (AR) and protein tyrosine phosphatase 1B (PTP1B), two enzymes strictly implicated in the development of DM and its complications, we synthesised 3-(5-arylidene-4-oxothiazolidin-3-yl)propanoic acids and analogous 2-butenoic acid derivatives, with the aim of balancing the effectiveness of dual AR/PTP1B inhibitors which we had identified as designed multiple ligands (DMLs). Out of the tested compounds, 4f exhibited well-balanced AR/PTP1B inhibitory effects at low micromolar concentrations, along with interesting insulin-sensitizing activity in murine C2C12 cell cultures. The SARs here highlighted along with their rationalization by in silico docking experiments into both target enzymes provide further insights into this class of inhibitors for their development as potential DML antidiabetic candidates.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Mellitus/drug therapy , Enzyme Inhibitors , Hypoglycemic Agents , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Aldehyde Reductase/metabolism , Animals , Diabetes Mellitus/enzymology , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hep G2 Cells , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Ligands , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Structure-Activity Relationship
5.
Carbohydr Res ; 499: 108220, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33341220

ABSTRACT

Diabetes mellitus is a multifactorial disease, which is frequently complicated by the development of hyperglycaemia-induced chronic complications. The therapy of diabetes mellitus often requires combinations of two or more drugs in order both to control glycaemic levels and to prevent hyperglycaemia-induced dangerous affairs. The application of multi-target agents, which are able to control simultaneously several pathogenic mechanisms, represents a useful alternative and, in fact, their discovery is a pursued aim of the research. Some (5-arylidene-4-oxo-2-thioxothiazolidin-3-yl)acetic acids, which we had previously reported as inhibitors of selected enzymes critically implicated in diabetes mellitus, were tested against pancreatic α-amylase and intestinal α-glucosidase. These enzymes catalyse the hydrolysis of dietary oligo- and polysaccharides into monosaccharides and, consequently, are responsible for postprandial hyperglycaemia; therefore, their inhibition is one of the possible strategies to control glycaemic levels in diabetes mellitus. In addition, we investigated the aggregation tendency of the tested compounds, through direct and indirect methods, in order to evaluate the mechanism of their multiple action and discover if aggregation may contribute to the inhibition of the target enzymes. Overall, compounds 1, 3 and 4 exhibited the most favourable profile since they were shown to act as multi-target inhibitors of enzymes involved in pathways related to diabetes mellitus, without producing aggregates even at high micromolar concentrations and, therefore, can be promising agents for further developments.


Subject(s)
Diabetes Mellitus/drug therapy , Glycoside Hydrolase Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Pancreatic alpha-Amylases/antagonists & inhibitors , Thiazolidines/pharmacology , Diabetes Mellitus/metabolism , Glycoside Hydrolase Inhibitors/adverse effects , Glycoside Hydrolase Inhibitors/chemistry , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/chemistry , Ligands , Molecular Structure , Pancreatic alpha-Amylases/metabolism , Thiazolidines/adverse effects , Thiazolidines/chemistry
6.
Bioorg Chem ; 92: 103211, 2019 11.
Article in English | MEDLINE | ID: mdl-31491569

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) acts as a negative regulator of insulin and leptin signalling and is crucially involved in the development of type 2 diabetes mellitus, obesity, cancer and neurodegenerative diseases. Pursuing our efforts to identify PTP1B inhibitors endowed with drug-like properties, we designed and evaluated 3-aryl-5-arylidene-2-thioxo-4-imidazolidinones (7) as a novel class of non-carboxylate PTP1B inhibitors. In agreement with our design, kinetic studies demonstrated that selected compounds 7 act as reversible, non-competitive inhibitors of the target enzyme at low micromolar concentrations. Accordingly, molecular docking experiments suggested that these inhibitors can fit an allosteric site of PTP1B that we previously individuated. Moreover, cellular assays demonstrated that compound 7e acts as a potent insulin-sensitizing agent in human liver HepG2 cells. Taken together, our results showed that these non-competitive PTP1B inhibitors can be considered promising lead compounds aimed to enhance druggability of the target enzyme and identify novel antidiabetic drugs.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Imidazolidines/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hep G2 Cells , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Imidazolidines/chemical synthesis , Imidazolidines/chemistry , Molecular Structure , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Bioorg Med Chem Lett ; 28(23-24): 3712-3720, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30342956

ABSTRACT

Designed multiple ligands (DMLs), developed to modulate simultaneously a number of selected targets involved in etiopathogenetic mechanisms of a multifactorial disease, such as diabetes mellitus (DM), are considered a promising alternative to combinations of drugs, when monotherapy results to be unsatisfactory. In this work, compounds 1-17 were synthesized and in vitro evaluated as DMLs directed to aldose reductase (AR) and protein tyrosine phosphatase 1B (PTP1B), two key enzymes involved in different events which are critical for the onset and progression of type 2 DM and related pathologies. Out of the tested 4-thiazolidinone derivatives, compounds 12 and 16, which exhibited potent AR inhibitory effects along with interesting inhibition of PTP1B, can be assumed as lead compounds to further optimize and balance the dual inhibitory profile. Moreover, several structural portions were identified as features that could be useful to achieve simultaneous inhibition of both human AR and PTP1B through binding to non-catalytic regions of both target enzymes.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Thiazolidines/chemistry , Thiazolidines/pharmacology , Aldehyde Reductase/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Ligands , Molecular Docking Simulation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 28(3): 278-283, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29292224

ABSTRACT

This paper describes the design, synthesis, and biological evaluation of 2-thioxoimidazolidin-4-one derivatives as inhibitors of proteasome and immunoproteasome, potential targets for the treatment of hematological malignancies. In particular, we focused our efforts on the design of noncovalent inhibitors, which might be a promising therapeutic option potentially devoid of drawbacks and side-effects related to irreversible inhibition. Among all the synthesized compounds, we identified a panel of active inhibitors with Ki values towards one or two chymotrypsin-like activities of proteasome (ß5c) and immunoproteasome (ß5i and ß1i subunits) in the low micromolar range. Docking studies suggested a unique binding mode of the molecules in the catalytic site of immunoproteasome proteolytic subunits.


Subject(s)
Imidazolidines/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Imidazolidines/chemical synthesis , Imidazolidines/chemistry , Molecular Docking Simulation , Molecular Structure , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry , Structure-Activity Relationship
9.
Sleep ; 40(9)2017 09 01.
Article in English | MEDLINE | ID: mdl-28651353

ABSTRACT

Study Objectives: Sleep fragmentation (SF) is highly prevalent and has emerged as an important contributing factor to obesity and metabolic syndrome. We hypothesized that SF-induced increases in protein tyrosine phosphatase-1B (PTP-1B) expression and activity underlie increased food intake, inflammation, and leptin and insulin resistance. Methods: Wild-type (WT) and ObR-PTP-1b-/- mice (Tg) were exposed to SF and control sleep (SC), and food intake was monitored. WT mice received a PTP-1B inhibitor (RO-7d; Tx) or vehicle (Veh). Upon completion of exposures, systemic insulin and leptin sensitivity tests were performed as well as assessment of visceral white adipose tissue (vWAT) insulin receptor sensitivity and macrophages (ATM) polarity. Results: SF increased food intake in either untreated or Veh-treated WT mice. Leptin-induced hypothalamic STAT3 phosphorylation was decreased, PTP-1B activity was increased, and reduced insulin sensitivity emerged both systemic and in vWAT, with the latter displaying proinflammatory ATM polarity changes. All of the SF-induced effects were abrogated following PTP-1B inhibitor treatment and in Tg mice. Conclusions: SF induces increased food intake, reduced leptin signaling in hypothalamus, systemic insulin resistance, and reduced vWAT insulin sensitivity and inflammation that are mediated by increased PTP-1B activity. Thus, PTP-1B may represent a viable therapeutic target in the context of SF-induced weight gain and metabolic dysfunction.


Subject(s)
Inflammation/metabolism , Insulin Resistance , Intra-Abdominal Fat/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Sleep Deprivation/physiopathology , Adipose Tissue, White/metabolism , Animals , Eating/physiology , Hypothalamus/metabolism , Inflammation/enzymology , Insulin/metabolism , Intra-Abdominal Fat/enzymology , Leptin/metabolism , Macrophages/metabolism , Male , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Receptor, Insulin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Sleep Deprivation/enzymology , Tyrosine/metabolism , Weight Gain
10.
Eur J Med Chem ; 127: 840-858, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27842892

ABSTRACT

New 4-{[5-arylidene-2-(4-fluorophenylimino)-4-oxothiazolidin-3-yl]methyl}benzoic acids (5) and 2-thioxo-4-thiazolidinone analogues (6) were synthesised as a part of a continuing search for new inhibitors of protein tyrosine phosphatase 1B (PTP1B), an enzyme which is implicated in metabolic disorders and inflammatory signaling. Most of the tested compounds were shown to be potent PTP1B inhibitors. Moreover, their inhibition mechanism was markedly influenced by the substituents in the positions 2 and 5, as kinetic studies indicated. Docking experiments suggested that certain derivatives 5 and 6 may efficiently fit into an allosteric site positioned between the ß-sheet including Leu71 and Lys73 and a lipophilic pocket closed by the loop consisting of Pro210 to Leu 204. In cellular assays, several of these new 4-thiazolidinone derivatives showed insulinomimetic and anti-inflammatory properties. Out of them, compound 5b exhibited the most promising profile, being able to promote the activation of both insulin receptor and downstream Akt protein as well as to increase 2-deoxyglucose cellular uptake. Interestingly, compound 5b was also able to interrupt critical events in inflammatory signaling.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Benzoic Acid/chemistry , Benzoic Acid/pharmacology , Insulin/metabolism , Peptidomimetics/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Allosteric Regulation/drug effects , Anti-Inflammatory Agents/chemical synthesis , Benzoic Acid/metabolism , Computer Simulation , Drug Design , Hep G2 Cells , Humans , Kinetics , Peptidomimetics/chemistry , Peptidomimetics/metabolism , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
11.
J Med Chem ; 58(5): 2047-67, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25375908

ABSTRACT

Aldose reductase (AR) is an aldo-keto reductase that has been widely investigated as an enzyme crucially involved in the pathogenesis of chronic complications associated with diabetes mellitus. Recently it was established that AR also acts as a key mediator of certain oxidative and inflammatory signaling pathways that are involved in the development of different human pathologies, such as cardiovascular disorders, sepsis, and cancer. These findings have renewed interest in the search for new AR inhibitors (ARIs) with improved profiles as potential therapeutic agents. In this review, recent advances in the field and promising future directions for developing ARIs are discussed.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Complications/drug therapy , Diabetes Mellitus/drug therapy , Enzyme Inhibitors/therapeutic use , Inflammation/drug therapy , Aldo-Keto Reductases , Humans
12.
Eur J Med Chem ; 81: 1-14, 2014 Jun 23.
Article in English | MEDLINE | ID: mdl-24819954

ABSTRACT

A series of 5-(carbamoylmethoxy)benzylidene-2-oxo/thioxo-4-thiazolidinone derivatives (6-9) were synthesized as inhibitors of aldose reductase (AR), enzyme which plays a crucial role in the development of diabetes complications as well as in the inflammatory processes associated both to diabetes mellitus and to other pathologies. In vitro inhibitory activity indicated that compounds 6-9a-d were generally good AR inhibitors. Acetic acid derivatives 8a-d and 9a-d were shown to be the best enzyme inhibitors among the tested compounds endowed with significant inhibitory ability levels reaching submicromolar IC50 values. Moreover, some representative AR inhibitors (7a, 7c, 9a, 9c, 9d) were assayed in cultures of human keratinocytes in order to evaluate their capability to reduce NF-kB activation and iNOS expression. Compound 9c proved to be the best derivative endowed with both interesting AR inhibitory effectiveness and ability to reduce NF-kB activation and iNOS expression. Molecular docking and molecular dynamics simulations were undertaken to investigate the binding modes of selected compounds into the active site of AR in order to rationalize the inhibitory effectiveness of these derivatives.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Models, Molecular , Thiazolidines/pharmacology , Aldehyde Reductase/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Lens, Crystalline/enzymology , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Structure-Activity Relationship , Thiazolidines/chemical synthesis , Thiazolidines/chemistry
13.
Eur J Med Chem ; 71: 112-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24287560

ABSTRACT

Insulin resistance is a complex altered metabolic condition characterized by impaired insulin signaling and implicated in the pathogenesis of serious human diseases, such as diabetes, obesity, neurodegenerative pathologies. In pursuing our aim to identify new agents able to improve cellular insulin sensitivity, we have synthesized new 4-[(5-arylidene-4-oxo-2-phenylimino/oxothiazolidin-3-yl)methyl]benzoic acids (5, 8) and evaluated their inhibitory activity towards human protein tyrosine phosphatases PTP1B, LMW-PTP and TCPTP, enzymes which are involved in the development of insulin resistance. Compounds 5 and 8 showed from moderate to significant selectivity toward PTP1B over both the highly homologous TCPTP and the two isoforms of human LMW-PTP. In addition, most of the tested compounds selectively inhibited LMW-PTP IF1 over the isoform IF2. Docking studies into the active sites of PTP1B and LMW-PTP aided the rationalization of the observed PTP inhibitory profile. Moreover, most tested compounds were capable to induce the insulin metabolic pathway in mouse C2C12 skeletal muscle cells by remarkably stimulating both IRß phosphorylation and 2-deoxyglucose cellular uptake.


Subject(s)
Benzoic Acid/chemistry , Benzoic Acid/pharmacology , Insulin/metabolism , Muscle, Skeletal/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Benzoic Acid/chemical synthesis , Cell Line , Humans , Insulin Resistance , Mice , Molecular Docking Simulation , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Structure-Activity Relationship
14.
Med Chem ; 9(1): 84-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22762165

ABSTRACT

5-Arylidene-2-oxo-4-thiazolidinones and 2-phenylimino analogues were evaluated for their antidegenerative activity on human chondrocyte cultures stimulated by IL-1ß and for their inhibitory capability against matrix metalloproteinase- 13. Our results indicated that 5-arylidene-4-thiazolidinone derivatives 1-9 exhibit antidegenerative activity and could block multiple cartilage destruction during the osteoarthritic process. Out of the selected compounds, (5-arylidene- 2,4-dioxothiazolidin-3-yl)acetic acids 7-9 showed significant effectiveness in reducing NO release and restoring normal levels of GAGs in chondrocytes treated with IL-1ß. Moreover, benzoic acids 1, 5 and 6 proved to be effective MMP-13 inhibitors and were able to restore normal levels of GAGs.


Subject(s)
Chondrocytes/drug effects , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinase Inhibitors/pharmacology , Thiazolidinediones/chemistry , Thiazolidinediones/pharmacology , Benzoates/chemistry , Benzoates/pharmacology , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Matrix Metalloproteinase 13/metabolism , Molecular Structure
15.
Eur J Med Chem ; 50: 332-43, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22381357

ABSTRACT

In pursuing our research targeting the identification of potent inhibitors of PTP1B and LMW-PTP, we have identified new 4-[(5-arylidene-2-arylimino-4-oxo-3-thiazolidinyl)methyl]benzoic acids endowed with interesting in vitro inhibitory profiles. Most compounds proved to be inhibitors of PTP1B and LMW-PTP isoform IF1. The tested inhibitors also showed selectivity towards PTP1B over the closely related TC-PTP. These compounds were found to activate the insulin-mediated signalling on mouse C2C12 skeletal muscle cells by increasing the phosphorylation levels of the insulin receptor and promoting cellular 2-deoxyglucose uptake. Interestingly, 4-{[5-(4-benzyloxybenzylidene)-2-(4-trifluoromethylphenylimino)-4-oxo-3-thiazolidinyl]methyl}benzoic acid (7d), the best in vitro inhibitor of PTP1B and the isoform IF1 of LMW-PTP, provided the highest activation level of the insulin receptor and was found to be endowed with an excellent insulinomimetic effect on the selected cells. This compound therefore represents an interesting lead compound for developing novel PTP1B and LMW-PTP inhibitors which could be achieved by improving both its pharmacological profile and its potentiating effects on insulin signalling.


Subject(s)
Benzoates/pharmacology , Biomimetics , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Muscle, Skeletal/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Thiazolidines/pharmacology , Animals , Benzoates/chemical synthesis , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/chemical synthesis , Humans , Mice , Models, Chemical , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Protein Conformation , Receptor, Insulin/metabolism , Thiazolidines/chemical synthesis , Tyrosine/metabolism
17.
Eur J Med Chem ; 46(7): 2797-806, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21531055

ABSTRACT

In continuing the search for more effective 5-arylidene-4-thiazolidinones as aldose reductase inhibitors, a new set of suitably substituted compounds (4, 5 and 8) was explored. Acetic acids 5, particularly 5a and 5h, proved to be interesting inhibitors of the enzyme as well as excellent antioxidant agents that are potentially able to counteract the oxidative stress associated with both diabetic complications as well as other pathologies. Molecular docking experiments supported SAR studies.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Antioxidants/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Thiazolidinediones/chemical synthesis , Acetates/chemistry , Aldehyde Reductase/chemistry , Animals , Antioxidants/pharmacology , Cattle , Diabetes Complications/prevention & control , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Oxidative Stress/drug effects , Structure-Activity Relationship , Thiazolidinediones/pharmacology
18.
Bioorg Med Chem Lett ; 21(1): 200-3, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21129963

ABSTRACT

2-Thioxo-4-thiazolidinone derivatives were evaluated as aldose reductase inhibitors (ARIs) and most of them exhibited good or excellent in vitro efficacy. Out of the tested compounds, most N-unsubstituted analogues were found to possess inhibitory effects at low micromolar doses and two of them exhibited higher potency than sorbinil, used as a reference drug. The insertion of an acetic chain on N-3 of the thiazolidinone scaffold led to analogues with submicromolar affinity for ALR2 and IC(50) values very similar to that of epalrestat, the only ARI currently used in therapy.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Thiazolidinediones/chemistry , Aldehyde Reductase/metabolism , Animals , Cattle , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Imidazolidines/chemistry , Imidazolidines/pharmacology , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacology
19.
Bioorg Med Chem ; 18(11): 4049-55, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20452228

ABSTRACT

Non-carboxylic acid containing bioisosteres of (5-arylidene-2,4-dioxothiazolidin-3-yl)acetic acids, which are active as aldose reductase (ALR2) inhibitors, were designed by replacing the carboxylic group with the trifluoromethyl ketone moiety. The in vitro evaluation of the ALR2 inhibitory effects of these trifluoromethyl substituted derivatives led to the identification of two inhibitors effective at low micromolar doses. It was further confirmed that a carboxylic chain on N-3 of the thiazolidinedione scaffold is a determining requisite to obtain the highest efficacy levels; however, it is not essential for the interaction with the target enzyme and it can be replaced by different polar groups, thus obtaining less ionised or unionised inhibitors.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Ketones/chemical synthesis , Animals , Chlorofluorocarbons, Methane , Humans , Ketones/pharmacology , Structure-Activity Relationship , Thiazolidinediones
20.
Eur J Med Chem ; 45(3): 1140-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20036445

ABSTRACT

The structure of aldehyde reductase (ALR1) in ternary complex with the coenzyme NADPH and [5-(3-carboxymethoxy-4-methoxybenzylidene)-2,4-dioxothiazolidin-3-yl]acetic acid (CMD), a potent inhibitor of aldose reductase (ALR2), was determined at 1.99A resolution. The partially disordered inhibitor formed a tight network of hydrogen bonds with the active site residues (Tyr50 and His113) and coenzyme. Molecular modelling calculations and inhibitory activity measurements of CMD and [5-(3-hydroxy-4-methoxybenzylidene)-2,4-dioxothiazolidin-3-yl]acetic acid (HMD) indicated that pi-stacking interactions with several conserved active site tryptophan residues and hydrogen-bonding interactions with the non-conserved C-terminal residue Leu300 in ALR2 (Pro301 in ALR1) contributed to inhibitor selectivity. In particular for the potent inhibitor CMD, the rotameric state of the conserved residue Trp219 (Trp220 in ALR1) is important in forming a pi-stacking interaction with the inhibitor in ALR2 and contributes to the difference in the binding of the inhibitor to the enzymes.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Aldehyde Reductase/chemistry , Catalytic Domain , Crystallography, X-Ray , Hydrogen Bonding , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Thiazolidinediones/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...