Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 248
Filter
1.
Clin Exp Immunol ; 193(2): 160-166, 2018 08.
Article in English | MEDLINE | ID: mdl-29698559

ABSTRACT

The role of helminth treatment in autoimmune diseases is growing constantly. Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease with challenging treatment options. Tuftsin-phosphorylcholine (TPC) is a novel helminth-based compound that modulates the host immune network. This study was conducted to evaluate the potential value of TPC in ameliorating lupus nephritis in a murine model and specifically to compare the efficacy of TPC to the existing first-line therapy for SLE: corticosteroids (methylprednisolone). Lupus-prone NZBxW/F1 mice were treated with TPC (5 µg/mouse), methylprednisolone (MP; 5 mg/body weight) or phosphate-buffered saline (PBS) (control) three times per week once glomerulonephritis, defined as proteinuria of grade > 100 mg/dl, was established. Levels of anti-dsDNA autoantibodies were evaluated by enzyme-linked immunosorbent assay (ELISA), splenic cytokines were measured in vitro and the kidney microscopy was analysed following staining. TPC and MP treatments improved lupus nephritis significantly and prolonged survival in NZBxW/F1 mice. TPC-treated mice showed a significantly decreased level of proteinuria (P < 0·001) and anti-dsDNA antibodies (P < 0·001) compared to PBS-treated mice. Moreover, TPC and MP inhibited the production of the proinflammatory cytokines interferon IFN-γ, interleukin IL-1ß and IL-6 (P < 0·001) and enhanced expression of the anti-inflammatory cytokine IL-10 (P < 0·001). Finally, microscopy analysis of the kidneys demonstrated that TPC-treated mice maintained normal structure equally to MP-treated mice. These data indicate that the small molecule named TPC hinders lupus development in genetically lupus-prone mice equally to methylprednisolone in most of the cases. Hence, TCP may be employed as a therapeutic potential for lupus nephritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Helminths/immunology , Kidney/pathology , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/drug therapy , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use , Tuftsin/therapeutic use , Animals , Antibodies, Antinuclear/blood , Cytokines/metabolism , Disease Models, Animal , Drug Combinations , Female , Humans , Inflammation Mediators/metabolism , Kidney/drug effects , Methylprednisolone/therapeutic use , Mice , Mice, Inbred NZB , Phosphorylcholine/chemistry , Tuftsin/chemistry
2.
Clin Exp Immunol ; 184(1): 19-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26618631

ABSTRACT

Treatment with helminthes and helminthes ova improved the clinical symptoms of several autoimmune diseases in patients and in animal models. Phosphorylcholine (PC) proved to be the immunomodulatory molecule. We aimed to decipher the tolerogenic potential of tuftsin-PC (TPC), a novel helminth-based compound in collagen-induced arthritis (CIA) a mouse model of rheumatoid arthritis (RA). CIA DBA/1 mice were treated with TPC subcutaneously (5 µg/0.1 ml) or orally (250 µg/0.1 ml), starting prior to disease induction. The control groups were treated with PBS. Collagen antibodies were tested by enzyme-linked immunosorbent assay (ELISA), cytokine protein levels by ELISA kits and regulatory T (Treg ) and regulatory B (Breg ) cell phenotypes by fluorescence-activated cell sorter (FACS). TPC-treated mice had a significantly lower arthritis score of 1.5 in comparison with control mice 11.8 (P < 0.0001) in both subcutaneous and orally treated groups at day 31. Moreover, histology analysis demonstrated highly inflamed joints in control mice, whereas TPC-treated mice maintained normal joint structure. Furthermore, TPC decreased the titres of circulating collagen II antibodies in mice sera (P < 0.0001), enhanced expression of IL-10 (P < 0.0001) and inhibited production of tumour necrosis factor (TNF)-α, interleukin (IL)-17 and IL-1ß (P < 0.0001). TPC significantly expanded the CD4(+) CD25(+) forkhead box protein 3 (FoxP3(+) ) Treg cells and CD19(+) IL-10(+) CD5(high) CD1d(high) T cell immunoglobulin mucin-1 (TIM-1(+) ) Breg cell phenotypes (P < 0.0001) in treated mice. Our data indicate that treatment with TPC attenuates CIA in mice demonstrated by low arthritic score and normal joints histology. TPC treatment reduced proinflammatory cytokines and increased anti-inflammatory cytokine expression, as well as expansion of Treg and Breg cells. Our results may lead to a new approach for a natural therapy for early rheumatoid arthritis onset.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies/blood , Arthritis, Experimental/drug therapy , Phosphorylcholine/pharmacology , Tuftsin/pharmacology , Administration, Oral , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/pathology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Collagen Type II/blood , Collagen Type II/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Hepatitis A Virus Cellular Receptor 1 , Immunophenotyping , Injections, Subcutaneous , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Joints/drug effects , Joints/immunology , Joints/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred DBA , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
Mini Rev Med Chem ; 12(5): 364-70, 2012 May.
Article in English | MEDLINE | ID: mdl-22303968

ABSTRACT

Chelators hold great promise as disease-modifying drugs for Alzheimer's therapy, and recent research efforts have focused on designing multi-target chelators with increased targeting and efficacy through rational drug design. In this review, we discuss our research studies on the rational design of new multi-target chelators with the potential not only to simultaneously modulate several disease-related targets, but also contain features designed to improve the BBB permeability, increase the brain targeting, and minimize potential side effects. These new chelators include neuroprotective chelators with brain selective monoamine oxidase (MAO) A/B inhibitory activity, acetylcholinesterase (AChE) inhibitors with site-activated chelating and neurogenesis activity, and AChE-MAO A/B inhibitors with site-activated chelating and neurogenesis activity.


Subject(s)
Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Antioxidants/pharmacology , Chelating Agents/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Monoamine Oxidase/metabolism , Alzheimer Disease/drug therapy , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Chelating Agents/chemistry , Chelating Agents/metabolism , Humans
4.
J Control Release ; 142(2): 214-20, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-19883707

ABSTRACT

Here we describe the design and application of OSu-FMS-MAL-S-(CH(2))(15)-COOH, an agent that associates with albumin while linked to a peptide or a protein with sufficient affinity (Ka=2 to 2.6 x 10(5)M(-1)) to protract the action of short- lived peptides and proteins in vivo. Under physiological conditions this probe undergoes spontaneous hydrolysis with the concomitant reactivation of inactive conjugates. Intravenously administered (125)I-labeled-Insulin-FMS-MAL-S-(CH(2))(15)-COOH to rats shows half-life of 17+/-2h, exceeding 5.2 times that obtained with intravenously administered (125)I-labeled Insulin. In mice this derivative facilitates glucose-lowering effect over a period of 24h, yielding AUC five times greater than that obtained by a similar dose of insulin-detemir. Similarly, subcutaneous administration of Exendin-4-FMS-MAL-S-(CH(2))(15)-COOH into mice facilitated prolonged and stable reduction in glucose level, yielding a t(1/2) value of 28+/-2h, exceeding the effect of exendin-4 4.7 folds. The inactive derivative gentamicin-FMS-MAL-S-(CH(2))(15)-COOH regained its full antibacterial potency upon incubation at physiological conditions yielding a t(1/2) value of 7.1+/-0.2h. In conclusion, the albumin-binding probe we introduced enables to prolong the action of any amino containing molecule in vivo, without the drawback of inactivation that often occurs upon such derivatization.


Subject(s)
Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Peptides/administration & dosage , Prodrugs/administration & dosage , Serum Albumin/metabolism , Venoms/administration & dosage , Amino Acid Sequence , Animals , Exenatide , Glucose/metabolism , Humans , Hydrolysis , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/chemistry , Insulin/metabolism , Insulin/therapeutic use , Mice , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Peptides/therapeutic use , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/therapeutic use , Protein Binding , Rats , Venoms/chemistry , Venoms/metabolism , Venoms/therapeutic use
5.
Eur J Pharm Biopharm ; 70(1): 19-28, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18495444

ABSTRACT

We attempted to engineer a novel long-acting insulin based on the following properties: (i) action as a prodrug to preclude supraphysiological concentrations shortly after injection; (ii) maintenance of low-circulating level of biologically active insulin for prolonged period; and (iii) high solubility in aqueous solution. A spontaneously hydrolyzable prodrug was thus designed and prepared by conjugating insulin through its amino side chains to a 40kDa polyethylene glycol containing sulfhydryl moiety (PEG(40)-SH), employing recently developed hetero-bifunctional spacer 9-hydroxymethyl-7(amino-3-maleimidopropionate)-fluorene-N-hydroxysucinimide (MAL-Fmoc-0Su). A conjugate trapped in the circulatory system and capable of releasing insulin by spontaneous chemical hydrolysis has been created. PEG(40)-Fmoc-insulin is a water-soluble, reactivatable prodrug with low biological activity. Upon incubation at physiological conditions, the covalently linked insulin undergoes spontaneous hydrolysis at a slow rate and in a linear fashion, releasing the nonmodified immunologically and biologically active insulin with a t(1/2) value of 30h. A single subcutaneous administration of PEG(40)-Fmoc-insulin to healthy and diabetic rodents facilitates prolonged glucose-lowering effects 4- to 7-fold greater than similar doses of the native hormone. The beneficial pharmacological features endowed by PEGylation are thus preserved. In contrast, nonreversible, "conventional" pegylation of insulin led to inactivation of the hormone.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Polyethylene Glycols/chemistry , Prodrugs/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cells, Cultured , Chemistry, Pharmaceutical , Delayed-Action Preparations , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Fluorenes/chemistry , Humans , Hydrolysis , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Injections, Intravenous , Injections, Subcutaneous , Insulin/administration & dosage , Insulin/chemistry , Insulin/pharmacokinetics , Lipogenesis/drug effects , Male , Maleimides/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats , Rats, Wistar , Time Factors
6.
Br J Cancer ; 97(12): 1655-63, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18071348

ABSTRACT

D(b-/-)xbeta2 microglobulin (beta2m) null mice transgenic for a chimeric HLA-A2.1/D(b)-beta2m single chain (HHD mice) are an effective biological tool to evaluate the antitumour cytotoxic T-lymphocyte response of known major histocompatibility-restricted peptide tumour-associated antigens, and to screen for putative unknown novel peptides. We utilised HHD lymphocytes to identify immunodominant epitopes of colon carcinoma overexpressed genes. We screened with HHD-derived lymphocytes over 500 HLA-A2.1-restricted peptides derived from colon carcinoma overexpressed genes. This procedure culminated in the identification of seven immunogenic peptides, three of these were derived from the 'human 1-8D gene from interferon inducible gene' (1-8D). The 1-8D gene was shown to be overexpressed in fresh tumour samples. The three 1-8D peptides were both antigenic and immunogenic in the HHD mice. The peptides induce cytotoxic T lymphocytes that were able to kill a colon carcinoma cell line HCT/HHD, in vitro and retard its growth in vivo. One of the peptides shared by all the 1-8 gene family primed efficiently normal human cytotoxic T lymphocyte precursors. These results highlight the 1-8D gene and its homologues as putative immunodominant tumour-associated antigens of colon carcinoma.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Interferons/chemistry , Interferons/pharmacology , Membrane Proteins/genetics , Peptides/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cross Reactions , Humans , Immunodominant Epitopes , Membrane Proteins/immunology , Mice , Mice, Transgenic
7.
J Neural Transm Suppl ; (70): 447-56, 2006.
Article in English | MEDLINE | ID: mdl-17017567

ABSTRACT

Iron and monoamine oxidase activity are increased in brain of Parkinson's disease (PD). They are associated with autoxidation and oxidative deamination of dopamine by MAO resulting in the generation of reactive oxygen species and the onset of oxidative stress to induce neurodegeneration. Iron chelators (desferal, Vk-28 and clioquinol) but not copper chelators have been shown to be neuroprotective in the 6-hydroxydoapmine and MPTP models of Parkinson's disease (PD), as are monoamine oxidase B inhibitors such as selegiline and rasagiline. These findings prompted the development of multifunctional anti PD drugs possessing iron chelating phamacophore of VK-28 and the propargylamine MAO inhibitory activity of rasagiline. M30 is a potent iron chelator, radical scavenger and brain selective irreversible MAO-A and B inhibitor, with little inhibition of peripheral MAO. It has neuroprotective activity in in vitro and in vivo models of PD and unlike selective MAO-B inhibitors it increases brain dopamine, serotonin and noradrenaline. These findings indicate beside its anti PD action, it may also possess antidepressant activity, similar to selective MAO-A and nonselective MAO inhibitors. These properties make it an ideal anti PD drug for which it is being developed.


Subject(s)
Hydroxyquinolines/therapeutic use , Iron Chelating Agents/therapeutic use , Monoamine Oxidase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Brain/drug effects , Brain/enzymology , Dopamine/physiology , Dopamine Agents/toxicity , Humans , Synaptic Transmission/drug effects
8.
Bioorg Khim ; 32(5): 459-66, 2006.
Article in Russian | MEDLINE | ID: mdl-17042263

ABSTRACT

Luliberin analogues modified at the N-terminus were synthesized to search for drugs exerting a cytotoxic effect on cells of hormone-dependent tumors. A synthetic scheme effective in the preparation of analogues containing fatty acid residues was proposed. The cytotoxic effect of the peptides was studied on a number of cell lines of human tumors in vitro. The dependence of the antitumor effect on the length of peptide chain, amino acid sequence, and structure of the N-terminal group was demonstrated. Modification with palmitic acid was found to result in highly active compounds in the case of analogues containing more than ten aa, whereas modifications with lauric, caproic, or trimethylacetic acid led to compounds with significantly lower activities. Analogues of luliberin containing a palmitic acid residue and effectively inhibiting the growth of tumor cells in vitro were synthesized.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Gonadotropin-Releasing Hormone/analogs & derivatives , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Drug Screening Assays, Antitumor , Gonadotropin-Releasing Hormone/chemistry , Humans , Molecular Sequence Data , Peptides/chemical synthesis
9.
J Neural Transm Suppl ; (71): 163-72, 2006.
Article in English | MEDLINE | ID: mdl-17447427

ABSTRACT

A large body of data indicates that a cascade of events contributes to the neurodegeneration in Alzheimer's disease (AD) and Parkinson's disease (PD). Metal (Fe, Cu, Zn) dyshomeostasis and oxidative stress are believed to play a pivotal role in the pathogenesis of these diseases. Accordingly, multifunctional compounds combining metal chelating and antioxidative activity hold a great promise as potential drugs for treating AD and PD. In this study, two novel NAPVSIPQ (NAP) analogs (M98 and M99) with potential antioxidant-metal chelating ability were designed and investigated, aiming to improve the poor metal chelating and antioxidative activity of NAP. Our studies showed that both M98 and M99 formed stable metal (Fe, Cu, Zn) complexes in water and demonstrated good metal (Fe, Cu, Zn) chelating properties as opposed to the poor metal (Fe, Cu, Zn) chelating properties of their parent peptide NAP. M98 and M99 exhibited significant inhibition of iron-induced lipid peroxidation in rat brain homogenates at concentrations of > or = 30 microM, while NAP failed to show any inhibition even at 100 microM. In human neuroblastoma cell (SH-SY5Y) culture, M98 and M99 at 1 microM completely protected against 6-hydroxydopamine (6OHDA) toxicity with potency similar to NAP and desferal (DFO), a strong iron chelator and a highly potent radical scavenger. In PC12 cell culture, M98 at the range of 0.001-1 microM displayed potent protection against 6-OHDA toxicity, comparable to NAP and DFO. These results suggest that M98 and M99 deserve further investigation as potential drug candidates for neuroprotection.


Subject(s)
Metals/toxicity , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Oxidative Stress/drug effects , Adrenergic Agents/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cell Survival/drug effects , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Humans , In Vitro Techniques , Oxidopamine/pharmacology , Rats , Siderophores/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism
10.
J Pept Res ; 66(4): 190-203, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16138857

ABSTRACT

A series of novel derivatives of neuropeptides with a metal-chelating moiety was synthesized and examined for various properties related to iron (Fe) chelation and neuroprotective action. All derivatives chelated Fe to form stable Fe complexes in water. Some strongly inhibited Fe-induced lipid peroxidation with an IC(50) value of about 12 microm. In PC12 cell culture, several compounds, at concentrations as low as 1 microm, attenuated serum-free stimulated cell death and improved cell survival by 20-35%. At this concentration, these analogs also protected against 6-hydroxydopamine (6-OHDA)-induced cell death, increasing cell viability by 20-30%. Electron paramagnetic resonance (EPR) studies indicated that besides being good Fe chelators, these analogs act as radical scavengers to directly scavenge hydroxyl radicals. Together, the data indicate that some of the analogs could be further developed as possible neuroprotective agents for treatment of neurodegenerative diseases such as Parkinson's, Alzheimer's, and Huntington's diseases, Friedreich's atxia, amyotrophic, and lateral sclerosis where Fe misregulation has been reported.


Subject(s)
Chelating Agents/chemical synthesis , Neurodegenerative Diseases/drug therapy , Neuropeptides/chemical synthesis , Animals , Chelating Agents/pharmacology , Free Radical Scavengers/metabolism , Metals, Heavy/metabolism , Neurodegenerative Diseases/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , PC12 Cells , Rats
11.
J Neural Transm (Vienna) ; 111(10-11): 1455-71, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15480846

ABSTRACT

Iron has been shown to accumulates at site where neurons degenerate in neurodegenerative diseases of Parkinson's disease, Alzheimer's disease, Huntington disease, amyotrophic lateral sclerosis and Friedreich ataxia. Iron is thought to participate or initiate oxidative stress via generation of reactive oxygen species (ROS), such as hydroxyl radical. Iron chelators are neuroprotective and prevent 6-hydroxydoapmine and MPTP dopaminergic neurotoxicity in rats and mice. However, their action on monoamine oxidase (MAO) A and B have not been determined previously since MAO-B inhibitors have been shown to be neuroprotective in cellular and animal models of Parkinson's disease. The chelators 8-hydroxyquinoline, O-phenanthroline, 2,2'-dipyridyl, U74500A and U74600F showed a preference for inhibition of rat brain mitochondrial MAO-A over MAO-B. Their IC(50) ranged from 10(-3) M to 10(-6) M, with 21-amino steroids (U74500A and U74006F) showing a greater selectivity and potency for MAO-A. Desferrioxamine (desferal), a prototype potent iron chelator, exhibited relatively poor MAO inhibitory. The inhibitions of MAO-A and B by 21-amino steroids (Lazaroids) were time dependent and irreversible. Those initiated by 8-hydroxyquinoline, 2,2'-dipyridyl and O-phenanthroline were fully reversible by enzyme dilution experiments. Both Fe(2+) and Fe(3+) reverse the MAO-A and B inhibition induced by the latter chelators, but not those initiated by 21-amino steroids. The data infer that either the inhibition of MAO by 21-amino steroids is either the resultant of their conversion to an irreversible covalently bound ligand or that the iron chelation moiety and MAO inhibitory activity in these compounds are not mutually shared. The results suggest that bifunctional brain penetrable drugs with iron chelating property and MAO inhibitory activity in could be the most feasible approach for neuroprotection in neurodegenerative diseases. Such drug would prevent participation of elevated iron in oxidative stress and formation of reactive hydroxyl radical, via its interaction with H(2)O2 (Fenton chemistry), generated as a consequence MAO and other oxidative enzyme reactions to generative cytotoxic reactive hydroxyl radical. We have now developed several of these compounds with neuroprotective, MAO inhibitory and iron chelating properties from our prototype iron chelators, VK-28 possessing propargylamine moiety of our anti-parkinson drug, rasagiline.


Subject(s)
Drug Delivery Systems , Iron Chelating Agents/therapeutic use , Monoamine Oxidase Inhibitors/therapeutic use , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Pargyline/analogs & derivatives , Parkinson Disease/drug therapy , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , In Vitro Techniques , Indans/therapeutic use , Male , Mitochondria/drug effects , Mitochondria/ultrastructure , Monoamine Oxidase/metabolism , Neurodegenerative Diseases/pathology , Pargyline/therapeutic use , Parkinson Disease/pathology , Piperazines/therapeutic use , Propylamines/therapeutic use , Quinolines/therapeutic use , Rats , Rats, Sprague-Dawley
12.
Biochem Biophys Res Commun ; 307(2): 315-21, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12859957

ABSTRACT

FMS(3)-insulin (2-sulfo-9-fluorenylmethoxycarbonyl)(3)-insulin is a water soluble, inactive-reactivated derivative of insulin with protracted action in vivo. In this study we find that FMS(3)-insulin preserves insulin's capacity to crystallize when associated with Zn(2+) ions or with basic protamine. Zinc or protamine suspended preparations of FMS(3)-insulin manifest substantially prolonged, blood glucose-lowering pharmacokinetic profiles in STZ-treated rats (STZ-rats). A dose of up to 1mg suspended FMS(3)-insulin/STZ rat can be subcutaneously administered with no hypoglycemic episodes at any time point after administration. This dose yielded glucose-lowering profiles with t(1/2) values at the range of 50-70h, turning catabolic STZ-rats into anabolic ones over a period of 2-3 days. The obtained glucose-lowering patterns exceeded 7-8 times in duration those produced by nonhypoglycemic doses of NPH-insulin. In summary, subcutaneous administration of suspended insulin prodrugs, such as FMS(3)-insulin, can bring about prolonged, nonhypoglycemic glucose-lowering profiles, unattainable with insulin preparations, which are known to be active at the time of administration.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Insulin/analogs & derivatives , Insulin/therapeutic use , Prodrugs/therapeutic use , Animals , Body Weight , Insulin/administration & dosage , Insulin/metabolism , Male , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/metabolism , Protamines/metabolism , Rats , Rats, Wistar , Streptozocin , Zinc/metabolism
13.
J Immunol ; 167(11): 6394-402, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11714805

ABSTRACT

The mast cell function-associated Ag (MAFA) is a type II membrane glycoprotein originally found on the plasma membrane of rat mucosal-type mast cells (RBL-2H3 line). A C-type lectin domain and an immunoreceptor tyrosine-based inhibitory motif (ITIM) are located in the extracellular and intracellular domains of MAFA, respectively. MAFA clustering has previously been shown to suppress the secretory response of these cells to the FcepsilonRI stimulus. Here we show that the tyrosine of the ITIM undergoes phosphorylation, on MAFA clustering, that is markedly enhanced on pervanadate treatment of the cells. Furthermore, the Src homology 3 domain of the protein tyrosine kinase Lyn binds directly to a peptide containing nonphosphorylated MAFA ITIM and PAAP motif. Results of both in vitro and in vivo experiments suggest that Lyn is probably responsible for this ITIM phosphorylation, which increases the Src homology domain 2 (SH2) affinity of Lyn for the peptide. In vitro measurements established that tyrosine-phosphorylated MAFA ITIM peptides also bind the SH2 domains of inositol 5'-phosphatase (SHIP) as well as protein tyrosine phosphatase-2. However, the former single domain is bound 8-fold stronger than both of the latter. Further support for the role of SHIP in the action of MAFA stems from in vivo experiments in which tyrosine-phosphorylated MAFA was found to bind primarily SHIP. In RBL-2H3 cells overexpressing wild-type SHIP, MAFA clustering causes markedly stronger inhibition of the secretory response than in control cells expressing normal SHIP levels or cells overexpressing either wild-type protein tyrosine phosphatase-2 or its dominant negative form. In contrast, on overexpression of the SH2 domain of SHIP, the inhibitory action of MAFA is essentially abolished. Taken together, these results suggest that SHIP is the primary enzyme responsible for mediating the inhibition by MAFA of RBL-2H3 cell response to the FcepsilonRI stimulus.


Subject(s)
Immunosuppressive Agents/metabolism , Lectins, C-Type , Mast Cells/enzymology , Mast Cells/immunology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Phosphoric Monoester Hydrolases/physiology , src Homology Domains/immunology , Amino Acid Sequence , Animals , Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Molecular Sequence Data , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Protein Binding/immunology , Rats , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/physiology , Tumor Cells, Cultured , Tyrosine/metabolism , Vanadates/pharmacology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , beta-N-Acetylhexosaminidases/metabolism , src Homology Domains/drug effects , src-Family Kinases/metabolism
14.
Neuron ; 32(2): 265-75, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11683996

ABSTRACT

We have determined the crystal structure at 1.8 A resolution of a complex of alpha-bungarotoxin with a high affinity 13-residue peptide that is homologous to the binding region of the alpha subunit of acetylcholine receptor. The peptide fits snugly to the toxin and adopts a beta hairpin conformation. The structures of the bound peptide and the homologous loop of acetylcholine binding protein, a soluble analog of the extracellular domain of acetylcholine receptor, are remarkably similar. Their superposition indicates that the toxin wraps around the receptor binding site loop, and in addition, binds tightly at the interface of two of the receptor subunits where it inserts a finger into the ligand binding site, thus blocking access to the acetylcholine binding site and explaining its strong antagonistic activity.


Subject(s)
Bungarotoxins/metabolism , Crystallography, X-Ray , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Receptors, Cholinergic/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallization , Dimerization , Disulfides/chemistry , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Conformation , Receptors, Cholinergic/metabolism
15.
Peptides ; 22(10): 1675-81, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11587796

ABSTRACT

Polymyxin B (PMB) is a potent antibacterial lipopeptide composed of a positively charged cyclic peptide ring and a fatty acid containing tail. Polymyxin B nonapeptide (PMBN), the deacylated amino derivative of polymyxin B, is much less bactericidal but able to permeabilize the outer membrane of Gram-negative bacteria and to neutralize the toxic effects of lipopolysaccharide (LPS). In this study, we synthesized and evaluated the antibacterial and LPS neutralizing activities of four PMBN analogs modified at their N-terminal. Our results suggest that oligoalanyl substitutions of PMBN do not effect most of PMBN activities. However, a hydrophobic aromatic substitution generated a PMB-like molecule with high antibacterial activity and significant reduced toxicity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides/metabolism , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Amino Acid Motifs , Animals , Anti-Bacterial Agents/chemistry , Binding Sites/physiology , Cells, Cultured/metabolism , Male , Mice , Microbial Sensitivity Tests , Polymyxin B/chemistry , Polymyxin B/isolation & purification
16.
Breast Cancer Res Treat ; 68(1): 55-64, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11678309

ABSTRACT

The effects of vasoactive intestinal peptide (VIP) antagonists on breast cancer cells were investigated. (N-stearyl, norleucine17)VIP hybrid ((SN)VIPhyb) inhibited specific 125I-VIP binding to MCF7, SKBR3, T47D ZR75-1 and MDA-MB231 cells with high affinity (IC50 values of 0.03-0.06 microM). (SN)VIPhyb, 1 microM, inhibited the ability of 10 nM VIP to cause elevation of cAMP and to increase c-fos mRNA. Micromolar concentrations of (SN)VIPhyb inhibited the proliferation of MDA-MB231 or MCF7 cells using a MTT and clonogenic assay. Using a MTT assay, (SN)VIPhyb enhanced the ability of taxol and doxorubicin to inhibit breast cancer growth. Using nude mice bearing MDA-MB231 xenografts, VIPhyb potentiated the ability of taxol to inhibit proliferation. The results indicate that VIP receptor antagonists increase the ability of chemotherapeutic drugs to kill breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Neurotensin/pharmacology , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Cyclic AMP/metabolism , Disease Models, Animal , Doxorubicin/pharmacology , Drug Synergism , Female , Genes, fos/drug effects , Humans , Iodine Radioisotopes , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neurotensin/therapeutic use , Paclitaxel/pharmacology , Protein Binding/drug effects , RNA, Messenger/drug effects , Receptors, Vasoactive Intestinal Peptide/metabolism , Recombinant Fusion Proteins/therapeutic use , Thymidine , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Vasoactive Intestinal Peptide/chemistry , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/therapeutic use
17.
Cancer ; 92(8): 2172-80, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11596035

ABSTRACT

BACKGROUND: Vasoactive intestinal peptide (VIP) is one of several small neuropeptides that affect cancer growth. A lipophilic VIP analog, stearyl-Nle(17)-neuroten-sin(6-11)VIP(7-28) (SNH) that inhibited lung carcinoma growth has been described previously. The experiments performed were clonogenic assays in vitro and tumor xenografts in nude mice in vivo. These studies were now extended to colon carcinoma and to combination therapy with chemotherapeutic agents. METHODS: Assays were performed with cell lines, and tumor proliferation was assessed using the (3-[4,5-dimethylthiazol-2-yl-5]-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]-2H tetrazolium) (MTS) colorimetric assay for mitochondrial function of living cells. RESULTS: The lipophilic analog (SNH) enhanced the antiproliferative activity of diverse chemotherapeutic agents: doxorubicine (antibiotic); vinorelbine (vinca alkaloid, antimicrotubule formation); paclitaxel (antimicrotubule agent); gemcitabine (antimetabolite); irinotecan (topoisomerase I inhibitor); and cisplatin (platinum compound acting as an alkylating agent). In all cases, the antiproliferative effect of SNH and the chemotheraputic agent was at least additive and for some combinations and concentrations even synergistic. For example, 2 microM of the antagonist that produced a 15-20% growth inhibition in the nonsmall cell lung carcinoma cell line reduced the IC(50) by 2-4-fold for most of the chemotherapeutic agents tested. Higher analog concentrations were even more efficacious. Similar results were obtained with colon carcinoma cell lines. CONCLUSIONS: Chemotherapeutic treatment of advanced solid tumors, such as nonsmall cell lung carcinoma, colon carcinoma, or prostate carcinoma, achieves a response rate of between 10% and 30% with significant toxicity. Combination therapy with the lipophilic VIP analog SNH and the preferred chemotherapeutic agent may greatly enhance the response rate, and by permitting a dose reduction, should significantly reduce side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Growth Inhibitors/pharmacology , Neurotensin/pharmacology , Recombinant Fusion Proteins/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Synergism , Humans , Lung Neoplasms , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Stem Cell Assay , Vasoactive Intestinal Peptide/metabolism , Xenograft Model Antitumor Assays
18.
J Med Chem ; 44(22): 3645-52, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11606129

ABSTRACT

The design, synthesis, and biological evaluation of a gonadotropin-releasing hormone (GnRH) agonist, [D-Lys(6)(1,3,8-trihydroxy-6-carboxyanthraquinone)]GnRH ([D-Lys(6)(Emo)]GnRH), is described. Synthesis of this analogue was carried out in a homogeneous solution as well as on a polymer support. [D-Lys(6)(Emo)]GnRH was found to bind to rat pituitary GnRH receptors (IC(50) = 0.25 nM), to induce luteinizing hormone (LH) release (ED(50) = 27 pM), and to be devoid of any toxicity. This analogue also proved to be a very potent agonist in vivo and exhibited a prolonged bioactivity. Six hours after its administration to rats, LH levels were substantially higher than those of rats treated with a 10-fold higher dose of the parent peptide. Moreover, chronic treatment of adult male rats with [D-Lys(6)(Emo)]GnRH (0.1 nmol/rat) for one week resulted in a further decrease of the weight of the testes and prostate as compared to those of rats that were treated with a higher dose of [D-Lys(6)]GnRH (1 nmol/rat). The prolonged activity of [D-Lys(6)(Emo)]GnRH may be attributed to its emodic acid moiety, which enhances the binding affinity of the analogue to human serum albumin. Indeed, we found that emodic acid binds to human serum albumin almost completely at the examined range of concentrations.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/chemical synthesis , Animals , DNA Fragmentation , Drug Design , Female , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/pharmacology , Humans , In Vitro Techniques , Luteinizing Hormone/blood , Male , Mice , Organ Size , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Prostate/drug effects , Protein Binding , Radioligand Assay , Rats , Rats, Wistar , Receptors, LHRH/agonists , Receptors, LHRH/metabolism , Serum Albumin/chemistry , Testis/drug effects , Tumor Cells, Cultured
19.
Photochem Photobiol ; 74(2): 149-56, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11547548

ABSTRACT

The electron-transfer properties of the hypericin derivatives, dibromo-, hexaacetyl-, hexamethyl- and desmethylhypericin, were studied. Cyclovoltammetric measurements revealed that dibromo- and desmethylhypericin have almost the same redox potentials as the parent hypericin. Substitution of the hydroxyl groups by acetoxy leads to less negative E1/2 values, whereas methoxy substitution induces more negative values. Electron paramagnetic resonance (EPR)/electron nuclear double resonance/general TRIPLE spectroscopy and quantum mechanical calculations were used to establish the structure of the one-electron reduced stages of hypericin derivatives. Proton loss in the bay region, already demonstrated for hypericin, was also found for dibromo- and desmethylhypericin. The spin and charge of the radical ions are predominately confined to the central biphenoquinone moiety of the hypericin skeleton. Generation of the radical ions by in situ electrolysis indicates that the redox potentials of hypericin, dibromo- and desmethylhypericin, containing hydroxyls at the 1, 3, 4, 6, 8 and 13 positions, largely depend on the solvent. With phosphate-buffered saline (pH 7.4)/dimethylsulfoxide (DMSO) as the solvent the EPR spectra of the corresponding radical ions appear at markedly lower potentials than in pure DMSO and N,N'-dimethylformamide. However, this effect is not observable for hexaacetyl- and hexamethyl-hypericin-lacking hydroxyl groups. In all cases the EPR data and calculations revealed the presence of 7,14 tautomers.


Subject(s)
Perylene/analogs & derivatives , Perylene/chemistry , Anions/chemistry , Anthracenes , Electron Spin Resonance Spectroscopy , Electron Transport , Free Radicals/chemistry , In Vitro Techniques , Molecular Structure , Oxidation-Reduction , Photochemistry , Photosensitizing Agents/chemistry
20.
Photochem Photobiol ; 74(2): 226-36, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11547560

ABSTRACT

In an attempt to develop an efficient chemotherapeutic agent targeted at malignant cells that express receptors to gonadotropin releasing hormone (GnRH) we coupled [D-Lys6]GnRH covalently to an emodin derivative, i.e. emodic acid (Emo) to yield [D-Lys6(Emo)]GnRH. Emodin is a naturally occurring anthraquinone which is widely used as a laxative and has other versatile biological activities. Physico-chemical studies employing electron paramagnetic resonance and electrochemistry of the conjugate as well as the (Emo) moiety showed that these compounds could be easily reduced either chemically, photochemically or enzymatically to their corresponding semiquinones. In the presence of oxygen the semiquinones generated reactive oxygen species (ROS), mainly superoxide and hydroxyl radicals, which were detected by the spin trapping method. Moreover, upon irradiation with visible light these compounds produced ROS and a highly reactive excited triplet state of Emo, which by itself may cause the oxidation of certain electron acceptors such as amino acids and bases of nucleic acids. Thus, [D-Lys6]GnRH-photosensitizer conjugates may be potentially used for targeted photodynamic chemotherapy aimed at treating cancer cells that carry GnRH receptors. These conjugates may also induce cytotoxicity in the dark similar to common conventional chemotherapeutic agents. The peptidic moiety, [D-Lys6]GnRH, was found to be stable toward highly reactive ROS generated either from enzymatic reduction or upon photoirradiation. The physico-chemical properties of Emo were only marginally influenced by the peptidic [D-Lys6]GnRH carrier.


Subject(s)
Emodin/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Emodin/chemistry , Emodin/pharmacology , Free Radicals/chemistry , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Photochemistry , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Receptors, LHRH/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...