Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 382(2): 233-245, 2022 08.
Article in English | MEDLINE | ID: mdl-35680376

ABSTRACT

Diabetes is associated with increased cardiac injury and sudden death. Nicotinamide phosphoribosyltransferase (Nampt) is an essential enzyme for the NAD+ salvage pathway and is dysregulated in diabetes. Nampt activation results in rescued NADH/NAD+ ratios and provides pharmacological changes necessary for diabetic cardioprotection. Computer docking shows that 1-(3,6-Dibromo-carbazol-9-yl)-3-phenylamino-propan-2-ol (P7C3) allows for enhanced Nampt dimerization and association. To test the pharmacological application, we used male leptin receptor-deficient (db/db) mice and treated them with Nampt activator P7C3. The effects of 4-week P7C3 treatment on cardiac function were evaluated along with molecular signaling changes for phosphorylated protein kinase B (p-AKT), phosphorylated endothelial nitric oxide synthase (p-eNOS), and sirtuin 1 (SIRT1). The cardiac function evaluated by ECG and echocardiography were significantly improved after 4 weeks of P7C3 treatment. Biochemically, higher NADH/NAD+ ratios in diabetic hearts were rescued by P7C3 treatment. Moreover, activities of Nampt and SIRT1 were significantly increased in P7C3-treated diabetic hearts. P7C3 treatment significantly decreased the blood glucose in diabetic mice with 4-week treatment as noted by glucose tolerance test and fasting blood glucose measurements compared with vehicle-treated mice. P7C3 activated Nampt enzymatic activity both in vitro and in the 4-week diabetic mouse hearts, demonstrating the specificity of the small molecule. P7C3 treatment significantly enhanced the expression of cardioprotective signaling of p-AKT, p-eNOS, and Beclin 1 in diabetic hearts. Nampt activator P7C3 allows for decreased infarct size with decreased Troponin I and lactose dehydrogenase (LDH) release, which is beneficial to the heart. Overall, the present study shows that P7C3 activates Nampt and SIRT1 activity and decreases NADH/NAD+ ratio, resulting in improved biochemical signaling providing cardioprotection. SIGNIFICANCE STATEMENT: This study shows that 1-(3,6-Dibromo-carbazol-9-yl)-3-phenylamino-propan-2-ol (P7C3) is effective in treating diabetes and cardiovascular diseases. The novel small molecule is antiarrhythmic and improves the ejection fraction in diabetic hearts. The study successfully demonstrated that P7C3 decreases the infarct size in hearts during myocardial infarction and ischemia-reperfusion injury. Biochemical and cellular signaling show increased NAD+ levels, along with Nampt activity involved in upregulating protective signaling in the diabetic heart. P7C3 has high therapeutic potential for rescuing heart disease.


Subject(s)
Diabetes Mellitus, Experimental , Myocardial Infarction , Animals , Blood Glucose , Carbazoles , Cytokines/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Male , Mice , Myocardial Infarction/drug therapy , NAD/metabolism , Nicotinamide Phosphoribosyltransferase , Proto-Oncogene Proteins c-akt , Sirtuin 1/metabolism
2.
Molecules ; 26(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925418

ABSTRACT

Fatty acid amides are a diverse family of underappreciated, biologically occurring lipids. Herein, the methods for the chemical synthesis and subsequent characterization of specific members of the fatty acid amide family are described. The synthetically prepared fatty acid amides and those obtained commercially are used as standards for the characterization and quantification of the fatty acid amides produced by biological systems, a fatty acid amidome. The fatty acid amidomes from mouse N18TG2 cells, sheep choroid plexus cells, Drosophila melanogaster, Bombyx mori, Apis mellifera, and Tribolium castaneum are presented.


Subject(s)
Amides/chemistry , Fatty Acids/chemistry , Lipids/chemistry , Amides/chemical synthesis , Amides/metabolism , Animals , Bees/chemistry , Bombyx/chemistry , Cell Line , Drosophila melanogaster/chemistry , Fatty Acids/chemical synthesis , Fatty Acids/metabolism , Lipids/genetics , Mice , Sheep , Tribolium/chemistry
3.
Front Mol Biosci ; 8: 801749, 2021.
Article in English | MEDLINE | ID: mdl-35047560

ABSTRACT

The fatty acid amides are a family of lipids composed of two chemical moieties, a fatty acid and a biogenic amine linked together in an amide bond. This lipid family is structurally related to the endocannabinoid anandamide (N-arachidonoylethanolamine) and, thus, is frequently referred to as a family of endocannabinoid-related lipids. The fatty acid amide family is divided into different classes based on the conjugate amine; anandamide being a member of the N-acylethanolamine class (NAE). Another class within the fatty acid amide family is the N-acyl amino acids (NA-AAs). The focus of this review is a sub-class of the NA-AAs, the N-acyl aromatic amino acids (NA-ArAAs). The NA-ArAAs are not broadly recognized, even by those interested in the endocannabinoids and endocannabinoid-related lipids. Herein, the NA-ArAAs that have been identified from a biological source will be highlighted and pathways for their biosynthesis, degradation, enzymatic modification, and transport will be presented. Also, information about the cellular functions of the NA-ArAAs will be placed in context with the data regarding the identification and metabolism of these N-acylated amino acids. A review of the current state-of-knowledge about the NA-ArAAs is to stimulate future research about this underappreciated sub-class of the fatty acid amide family.

4.
Article in English | MEDLINE | ID: mdl-33284726

ABSTRACT

Octolig, a commercially available (a polyethylene diamine covalently attached to silica gel), was subjected to modifications to incorporate sulfur for enhanced removal of lead ion from aqueous solutions. The basic approach was attempted formation of "ThioOctolig" by the reaction of Octolig with thioacetamide in toluene using a shaker bath for 24 or 48 h or in the presence of 10% HCl (1 h). Our experience was that conversion was limited to about 20% based on sulfur analysis for 24 or 48 h reaction time, or in the presence of 10% HCl. In fact, with acidification, the results were poorer. Duplicate runs indicated consistent results. Literature reported that SbCl3 was an effective catalyst with a reaction time of 1 h. Use of this reagent (1-h reaction time) produced a bright orange red product, in contrast with previous yellow-colored products. A control run indicated that this reagent reacted with Octolig in toluene (in the dark) to produce a red-colored sample; thioacetamide reacted to produce a yellow sample. Use of SbCl3 (∼5 mole %) did not enhance the sulfur content of Octolig. A sample of Octolig removed 68% lead ion from a 120 ppm aqueous lead while a sample of ThioOctolig (10% S) removed 99.4% lead ions. We also investigated enhancing the sulfur incorporation upon raising the reaction temperature with thioacetamide.


Subject(s)
Chelating Agents/chemistry , Lead/analysis , Polyethylenes/chemistry , Silica Gel/chemistry , Thioacetamide/chemistry , Water Pollutants, Chemical/analysis , Adsorption , Chelating Agents/chemical synthesis , Drinking Water/analysis , Drinking Water/standards , Models, Theoretical , Surface Properties , Water Purification/methods
5.
J Org Chem ; 85(15): 9928-9935, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32674572

ABSTRACT

A family of three spatially directional resorcin[4]arene cavitand glycoconjugates (RCGs) have been applied as efficient recoverable and reusable inverse phase transfer catalysts for eco- and environmentally friendly thiocyanation and 2-amino-1,3-thiazole formation reactions in water. The results show that RCGs (1 mol %) were capable of hosting and catalyzing various water-insoluble bromo/thiocyanato substrates in water without the use of any co-organic solvents. The recoverability and reusability of RCG catalytic systems, that is, RCG1 and RCG3, were also examined upon a simple extraction of the desired products using DCM or ethyl acetate, followed by subjecting the recovered aqueous solution containing the RCG catalysts to the next reaction cycles.

6.
RSC Adv ; 9(18): 10109-10116, 2019 Mar 28.
Article in English | MEDLINE | ID: mdl-35520904

ABSTRACT

The Cu(i)-catalyzed azide-alkyne cycloaddition (CuAAC) in aqueous media using resorcin[4]arene glycoconjugate (RG) is reported. The eight ß-d-glucopyranoside moieties constructed on the resorcin[4]arene upper rim provide a pseudo-saccharide cavity that offers a suitable host environment for water-insoluble hydrophobic azido and/or alkyne substrates in water. The utility of RG was established as an efficient inverse phase transfer catalyst for the CuAAC in water as a green approach for the synthesis of 1,4-disubstituted 1,2,3-triazole species. The catalytic utility of RG (1 mol%) was demonstrated in a multicomponent one-pot CuAAC for various azido/alkyne substrates. The RG acts as a molecular host and a micro-reactor resulting in the 1,4-disubstituted 1,2,3-triazoles in excellent yield.

7.
Article in English | MEDLINE | ID: mdl-29775126

ABSTRACT

Aqueous Li+ - containing samples (in DI water or well water) were eluted over Octolig®, a polyethylenediimine covalently attached to a high- surface-area silica gel, and only slight removal, if any, could be claimed. However, when using tetrahydrofuran (THF) as a solvent we quantitatively removed lithium ion with Octolig® or with alkylated Octolig®, demonstrating the efficacy of Octolig® and lack of advantage of a N, N'-dialkylated Octolig®. In addition, the removal of alkali metal ions (lithium, sodium, and potassium) in THF by Octolig® was partially selective: While being quantitative for lithium it was only about 40% for potassium. The study has potential implications for using geothermal brines not only as a heat source, but as a source of lithium as well.


Subject(s)
Lithium Compounds/isolation & purification , Lithium/isolation & purification , Methane/analogs & derivatives , Water Pollutants, Chemical/isolation & purification , Water Purification/methods , Water/chemistry , Anions/isolation & purification , Lithium/chemistry , Methane/chemistry , Methane/isolation & purification , Silica Gel/chemistry , Water Purification/instrumentation
8.
Drug Deliv ; 24(1): 1317-1324, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28906151

ABSTRACT

Conventional therapeutic techniques treat patients by delivering biotherapeutics to the entire body. With targeted delivery, biotherapeutics are transported to the afflicted tissue reducing exposure to healthy tissue. Targeted delivery devices are minimally composed of a stimuli responsive polymer allowing triggered release and magnetic nanoparticles enabling targeting as well as alternating magnetic field (AMF) heating. Although more traditional methods, like emulsion polymerization, have been used to realize such devices, the synthesis is problematic. For example, surfactants preventing agglomeration must be removed from the product increasing time and cost. Ultraviolet (UV) photopolymerization is more efficient and ensures safety by using biocompatible substances. Reactants selected for nanogel fabrication were N-isopropylacrylamide (monomer), methylene bis-acrylamide (crosslinker), and Irgacure 2959 (photoinitiator). The 10 nm superparamagnetic nanoparticles for encapsulation were composed of iron oxide. Herein, a low-cost, scalable, and rapid, custom-built UV photoreactor with in situ, spectroscopic monitoring system is used to observe synthesis. This method also allows in situ encapsulation of the magnetic nanoparticles simplifying the process. Nanogel characterization, performed by transmission electron microscopy, reveals size-tunable nanogel spheres between 40 and 800 nm in diameter. Samples of nanogels encapsulating magnetic nanoparticles were subjected to an AMF and temperature increase was observed indicating triggered release is possible. Results presented here will have a wide range of applications in medical sciences like oncology, gene delivery, cardiology, and endocrinology.


Subject(s)
Metal Nanoparticles , Polyethylene Glycols , Polyethyleneimine , Acrylic Resins , Ferric Compounds , Nanogels
9.
Chemistry ; 22(18): 6223-7, 2016 Apr 25.
Article in English | MEDLINE | ID: mdl-26933945

ABSTRACT

The synthesis of novel spatially directional multivalent resorcin[4]arene cavitand glycoconjugates (RCGs) and their ability to catalyze organic reactions is reported. The ß-d-glucopyranoside moieties on the upper rim of the "bowl"-shaped resorcin[4]arene cavitand core are capable of multiple hydrogen-bond interactions resulting in a pseudo-cavity, which has been investigated for organic transformations in aqueous media. The RCGs have been demonstrated to catalyze thiazole formation, thiocyanation, copper(I)-catalyzed azide alkyne cycloaddition (CuAAC), and Mannich reactions; they impart stereoselectivity in the three-component Mannich reaction. Thermodynamic values obtained from (1) H diffusion-ordered spectroscopy (DOSY) experiments suggest that the upper saccharide cavity of the RCG and not the resorcin[4]arene cavity is the site of the complexation event.


Subject(s)
Alkynes/chemistry , Azides/chemistry , Calixarenes/chemistry , Glycoconjugates/chemistry , Phenylalanine/analogs & derivatives , Catalysis , Copper/chemistry , Cycloaddition Reaction , Phenylalanine/chemistry , Thermodynamics
10.
PLoS One ; 9(8): e105568, 2014.
Article in English | MEDLINE | ID: mdl-25162558

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and the most aggressive form of primary brain tumor. Jak2 is a non-receptor tyrosine kinase that is involved in proliferative signaling through its association with various cell surface receptors. Hyperactive Jak2 signaling has been implicated in numerous hematological disorders as well as in various solid tumors including GBM. Our lab has developed a Jak2 small molecule inhibitor known as G6. It exhibits potent efficacy in vitro and in several in vivo models of Jak2-mediated hematological disease. Here, we hypothesized that G6 would inhibit the pathogenic growth of GBM cells expressing hyperactive Jak2. To test this, we screened several GBM cell lines and found that T98G cells express readily detectable levels of active Jak2. We found that G6 treatment of these cells reduced the phosphorylation of Jak2 and STAT3, in a dose-dependent manner. In addition, G6 treatment reduced the migratory potential, invasive potential, clonogenic growth potential, and overall viability of these cells. The effect of G6 was due to its direct suppression of Jak2 function and not via off-target kinases, as these effects were recapitulated in T98G cells that received Jak2 specific shRNA. G6 also significantly increased the levels of caspase-dependent apoptosis in T98G cells, when compared to cells that were treated with vehicle control. Lastly, when T98G cells were injected into nude mice, G6 treatment significantly reduced tumor volume and this was concomitant with significantly decreased levels of phospho-Jak2 and phospho-STAT3 within the tumors themselves. Furthermore, tumors harvested from mice that received G6 had significantly less vimentin protein levels when compared to tumors from mice that received vehicle control solution. Overall, these combined in vitro and in vivo results indicate that G6 may be a viable therapeutic option against GBM exhibiting hyperactivation of Jak2.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Nude , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Small Molecule Libraries/chemical synthesis , Tumor Burden/drug effects , Vimentin/antagonists & inhibitors , Vimentin/genetics , Vimentin/metabolism , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 288(37): 26834-46, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23902767

ABSTRACT

Increased food intake and lack of physical activity results in excess energy stored in adipocytes, and this imbalance contributes to obesity. New adipocytes are required for storage of energy in the white adipose tissue. This process of adipogenesis is widely studied in differentiating 3T3L1 preadipocytes in vitro. We have identified a key signaling kinase, protein kinase C delta (PKCδ), whose alternative splice variant expression is modulated during adipogenesis. We demonstrate that PKCδII splice variant promotes survival in differentiating 3T3L1 cells through the Bcl2 pathway. Here we demonstrate that resveratrol, a naturally occurring polyphenol, increases apoptosis and inhibits adipogenesis along with disruption of PKCδ alternative splicing during 3T3L1 differentiation. Importantly, we have identified a PKCδII splice variant inhibitor. This inhibitor may be a valuable tool with therapeutic implications in obesity.


Subject(s)
Adipogenesis , Alternative Splicing , Apoptosis , Protein Kinase C-delta/antagonists & inhibitors , Stilbenes/chemistry , 3T3-L1 Cells , Animals , Cell Differentiation , Gene Expression Regulation, Enzymologic , Male , Mice , Mice, Inbred C57BL , Polyphenols/chemistry , Protein Kinase C-delta/genetics , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Resveratrol , Transfection
12.
Am J Pathol ; 181(3): 858-65, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22796437

ABSTRACT

Philadelphia chromosome-negative myeloproliferative neoplasms, including polycythemia vera, essential thrombocytosis, and myelofibrosis, are disorders characterized by abnormal hematopoiesis. Among these myeloproliferative neoplasms, myelofibrosis has the most unfavorable prognosis. Furthermore, currently available therapies for myelofibrosis have little to no efficacy in the bone marrow and hence, are palliative. We recently developed a Janus kinase 2 (Jak2) small molecule inhibitor called G6 and found that it exhibits marked efficacy in a xenograft model of Jak2-V617F-mediated hyperplasia and a transgenic mouse model of Jak2-V617F-mediated polycythemia vera/essential thrombocytosis. However, its efficacy in Jak2-mediated myelofibrosis has not previously been examined. Here, we hypothesized that G6 would be efficacious in Jak2-V617F-mediated myelofibrosis. To test this, mice expressing the human Jak2-V617F cDNA under the control of the vav promoter were administered G6 or vehicle control solution, and efficacy was determined by measuring parameters within the peripheral blood, liver, spleen, and bone marrow. We found that G6 significantly reduced extramedullary hematopoiesis in the liver and splenomegaly. In the bone marrow, G6 significantly reduced pathogenic Jak/STAT signaling by 53%, megakaryocytic hyperplasia by 70%, and the Jak2 mutant burden by 68%. Furthermore, G6 significantly improved the myeloid to erythroid ratio and significantly reversed the myelofibrosis. Collectively, these results indicate that G6 is efficacious in Jak2-V617F-mediated myelofibrosis, and given its bone marrow efficacy, it may alter the natural history of this disease.


Subject(s)
Janus Kinase 2/metabolism , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/enzymology , Protein Kinase Inhibitors/therapeutic use , Small Molecule Libraries/therapeutic use , Stilbenes/therapeutic use , Amino Acid Substitution/genetics , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Disease Models, Animal , Hematopoiesis, Extramedullary/drug effects , Humans , Hyperplasia , Janus Kinase 2/antagonists & inhibitors , Megakaryocytes/drug effects , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myeloid Cells/drug effects , Myeloid Cells/pathology , Phosphorylation/drug effects , Primary Myelofibrosis/blood , Primary Myelofibrosis/physiopathology , Protein Kinase Inhibitors/pharmacology , Reticulin/drug effects , Reticulin/metabolism , STAT5 Transcription Factor/metabolism , Small Molecule Libraries/pharmacology , Spleen/drug effects , Spleen/pathology , Spleen/physiopathology , Splenomegaly/complications , Splenomegaly/drug therapy , Splenomegaly/pathology , Splenomegaly/physiopathology , Stilbenes/pharmacology
13.
Bioorg Med Chem Lett ; 22(3): 1402-7, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22227213

ABSTRACT

In this study, we analyzed the structure-activity relationship properties of the small molecule Jak2 inhibitor G6. We synthesized a set of derivatives containing the native para-hydroxyl structure or an alternative meta-hydroxyl structure and examined their Jak2 inhibitory properties. We found that the para-hydroxyl derivative known as NB15 had excellent Jak2 inhibitory properties in silico, in vitro, and ex vivo when compared with meta-hydroxyl derivatives. These results indicate that NB15 is a potent derivative of the Jak2 inhibitor G6, and that maintaining the para-hydroxyl orientation of G6 is critical for its Jak2 inhibitory potential.


Subject(s)
Benzylamines/chemistry , Benzylamines/pharmacology , Janus Kinase 2/antagonists & inhibitors , Models, Molecular , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Hydroxylation , Mice , Mice, Transgenic , Molecular Structure , Stilbenes/chemistry , Stilbenes/pharmacology , Structure-Activity Relationship
14.
Exp Hematol ; 40(1): 22-34, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22019628

ABSTRACT

Hyperkinetic Jak2 tyrosine kinase signaling has been implicated in several hematological disorders, including myeloproliferative neoplasms. Effective Jak2 inhibitors can have significant therapeutic potential. Here, using structure-based virtual screening, we identified a benzothiophene-derived Jak2 inhibitor named A46. We hypothesized that this compound would inhibit Jak2-V617F-mediated pathologic cell growth. To test this, A46 was analyzed for its ability to inhibit recombinant Jak2 protein catalysis; suppress Jak2-mediated pathogenic cell growth in vitro; inhibit the aberrant ex vivo growth of Jak2-V617F-expressing primary human bone marrow cells; and inhibit Jak2-mediated pathogenesis in vivo. To this end, we found that A46 selectively inhibited Jak2-V617F protein when compared to wild-type Jak2 protein. The drug also selectively inhibited the proliferation of Jak2-V617F-expressing cells in both a time- and dose-dependent manner, and this correlated with decreased Jak2 and signal transducers and activators of transcription 5 phosphorylation within treated cells. The Jak2-V617F cell growth inhibition correlated with an induction of cell cycle arrest and promotion of apoptosis. A46 also inhibited the pathologic growth of primary Jak2-V617F-expressing bone marrow cells ex vivo. Lastly, using a mouse model of Jak2-V617F-mediated myeloproliferative neoplasia. A46 significantly reduced the splenomegaly and megakaryocytic hyperplasia in the spleens of treated mice and the levels of interleukin-6 in the plasma. Collectively, our data demonstrate that the benzothiophene-based compound, A46, suppresses Jak2-mediated pathogenesis, thereby making it a potential candidate drug against Jak2-mediated disorders.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Janus Kinase 2/antagonists & inhibitors , Thiophenes/pharmacology , Animals , Apoptosis/drug effects , Biocatalysis , Bone Marrow Cells/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Janus Kinase 2/metabolism , Mice , Mice, Transgenic , Structure-Activity Relationship , Thiophenes/chemistry , Tumor Cells, Cultured
15.
Neoplasia ; 13(11): 1058-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22131881

ABSTRACT

We recently developed a Janus kinase 2 (Jak2) small-molecule inhibitor called G6 and found that it inhibits Jak2-V617F-mediated pathologic cell growth in vitro, ex vivo, and in vivo. However, its ability to inhibit Jak2-V617F-mediated myeloproliferative neoplasia, with particular emphasis in the bone marrow, has not previously been examined. Here, we investigated the efficacy of G6 in a transgenic mouse model of Jak2-V617F-mediated myeloproliferative neoplasia. We found that G6 provided therapeutic benefit to the peripheral blood as determined by elimination of leukocytosis, thrombocytosis, and erythrocytosis. G6 normalized the pathologically high plasma concentrations of interleukin 6 (IL-6). In the liver, G6 eliminated Jak2-V617F-driven extramedullary hematopoiesis. With respect to the spleen, G6 significantly reduced both the splenomegaly and megakaryocytic hyperplasia. In the critically important bone marrow, G6 normalized the pathologically high levels of phospho-Jak2 and phospho-signal transducer and activator of transcription 5 (STAT5). It significantly reduced the megakaryocytic hyperplasia in the marrow and completely normalized the M/E ratio. Most importantly, G6 selectively reduced the mutant Jak2 burden by 67%on average, with virtual elimination of mutant Jak2 cells in one third of all treated mice. Lastly, clonogenic assays using marrow stem cells from the myeloproliferative neoplasm mice revealed a time-dependent elimination of the clonogenic growth potential of these cells by G6. Collectively, these data indicate that G6 exhibits exceptional efficacy in the peripheral blood, liver, spleen, and, most importantly, in the bone marrow, thereby raising the possibility that this compound may alter the natural history of Jak2-V617F-mediated myeloproliferative neoplasia.


Subject(s)
Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Stilbenes/therapeutic use , Amino Acid Substitution/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Neoplasms/pathology , Cells, Cultured , Drug Evaluation, Preclinical , Janus Kinase 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phenylalanine/genetics , Protein Kinase Inhibitors/pharmacology , Stilbenes/pharmacology , Valine/genetics
16.
J Biol Chem ; 286(6): 4280-91, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21127060

ABSTRACT

Using structure-based virtual screening, we previously identified a novel stilbenoid inhibitor of Jak2 tyrosine kinase named G6. Here, we hypothesized that G6 suppresses Jak2-V617F-mediated human pathological cell growth in vitro and in vivo. We found that G6 inhibited proliferation of the Jak2-V617F expressing human erythroleukemia (HEL) cell line by promoting marked cell cycle arrest and inducing apoptosis. The G6-dependent increase in apoptosis levels was concomitant with increased caspase 3/7 activity and cleavage of PARP. G6 also selectively inhibited phosphorylation of STAT5, a downstream signaling target of Jak2. Using a mouse model of Jak2-V617F-mediated hyperplasia, we found that G6 significantly decreased the percentage of blast cells in the peripheral blood, reduced splenomegaly, and corrected a pathologically low myeloid to erythroid ratio in the bone marrow by eliminating HEL cell engraftment in this tissue. In addition, drug efficacy correlated with the presence of G6 in the plasma, marrow, and spleen. Collectively, these data demonstrate that the stilbenoid compound, G6, suppresses Jak2-V617F-mediated aberrant cell growth. As such, G6 may be a potential therapeutic lead candidate against Jak2-mediated, human disease.


Subject(s)
Cell Cycle/drug effects , Janus Kinase 2/metabolism , Leukemia, Erythroblastic, Acute/enzymology , Mutation, Missense , Protein Kinase Inhibitors/pharmacology , Stilbenes/pharmacology , Amino Acid Substitution , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Disease Models, Animal , Humans , Janus Kinase 2/genetics , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/genetics , Mice , Mice, Mutant Strains , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
17.
J Biol Chem ; 285(41): 31399-407, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20667821

ABSTRACT

Somatic mutations in the Jak2 protein, such as V617F, cause aberrant Jak/STAT signaling and can lead to the development of myeloproliferative neoplasms. This discovery has led to the search for small molecule inhibitors that target Jak2. Using structure-based virtual screening, our group recently identified a novel small molecule inhibitor of Jak2 named G6. Here, we identified a structure-function correlation of this compound. Specifically, five derivative compounds of G6 having structural similarity to the original lead compound were obtained and analyzed for their ability to (i) inhibit Jak2-V617F-mediated cell growth, (ii) inhibit the levels of phospho-Jak2, phospho-STAT3, and phospho-STAT5; (iii) induce apoptosis in human erythroleukemia cells; and (iv) suppress pathologic cell growth of Jak2-V617F-expressing human bone marrow cells ex vivo. Additionally, we computationally examined the interactions of these compounds with the ATP-binding pocket of the Jak2 kinase domain. We found that the stilbenoid core-containing derivatives of G6 significantly inhibited Jak2-V617F-mediated cell proliferation in a time- and dose-dependent manner. They also inhibited phosphorylation of Jak2, STAT3, and STAT5 proteins within cells, resulting in higher levels of apoptosis via the intrinsic apoptotic pathway. Finally, the stilbenoid derivatives inhibited the pathologic growth of Jak2-V617F-expressing human bone marrow cells ex vivo. Collectively, our data demonstrate that G6 has a stilbenoid core that is indispensable for maintaining its Jak2 inhibitory potential.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Polycythemia Vera/drug therapy , Protein Kinase Inhibitors/pharmacology , Stilbenes/pharmacology , Amino Acid Substitution , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mutation, Missense , Polycythemia Vera/enzymology , Polycythemia Vera/genetics , Protein Kinase Inhibitors/chemistry , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Stilbenes/chemistry , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 19(13): 3598-601, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19447617

ABSTRACT

Janus kinase 2 (JAK2) plays a crucial role in the pathomechanism of myeloproliferative disorders and hematologic malignancies. A somatic mutation of JAK2 (Val617Phe) was previously shown to occur in 98% of patients with polycythemia vera and 50% of patients with essential thrombocythemia and primary myelofibrosis. Thus, effective JAK2 kinase inhibitors may be of significant therapeutic importance. Here, we applied a structure-based virtual screen to identify novel JAK2 inhibitors. One JAK2 inhibitor in particular, G6, demonstrated remarkable potency as well as specificity, which makes it as a potential lead candidate against diseases related to elevated JAK2 tyrosine kinase activity.


Subject(s)
Alkenes/chemistry , Janus Kinase 2/antagonists & inhibitors , Phenols/chemistry , Protein Kinase Inhibitors/chemistry , Alkenes/pharmacology , Catalytic Domain , Cell Line, Tumor , Computer Simulation , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Janus Kinase 2/metabolism , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Phenols/pharmacology , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
19.
Chem Commun (Camb) ; (14): 1822-4, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19319413

ABSTRACT

Highly crystalline directional poly(epsilon-caprolactone) based on a tetrahydroxymethyl resorcin[4]arene initiator core was synthesized by a "core first" method via ring-opening polymerization catalyzed by Sn(Oct)(2) in bulk at 120 degrees C.


Subject(s)
Calixarenes/chemistry , Ethers, Cyclic/chemistry , Phenylalanine/analogs & derivatives , Polyesters/chemistry , Resorcinols/chemistry , Caprylates/chemistry , Catalysis , Phenylalanine/chemistry , Polyesters/chemical synthesis
20.
Biomacromolecules ; 9(10): 2921-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18771312

ABSTRACT

Enantiomerically pure functional polycarbonate was synthesized from a novel seven-membered cyclic carbonate monomer derived from naturally occurring L-tartaric acid. The monomer was synthesized in three steps and screened for polymerization with four commercially available lipases from different sources at 80 degrees C, in bulk. The ring-opening polymerization (ROP) was affected by the source of the enzyme; the highest number-average molecular weight, M(n) = 15500 g/mol (PDI = 1.7; [alpha]D(20) = +77.8, T(m) = 58.8 degrees C) optically active polycarbonate was obtained with lipase Novozyme-435. The relationship between monomer conversion, reaction time, molecular weight, and molecular weight distribution were investigated for Novozyme-435 catalyzed ROP. Deprotection of the ketal groups was achieved with minimal polymer chain cleavage (M(n) = 10000 g/mol, PDI = 2.0) and resulted in optically pure polycarbonate ([alpha]D(20) = +56) bearing hydroxy functional groups. Deprotected poly(ITC) shows T(m) of 60.2 degrees C and DeltaH(f) = 69.56 J/g and similar to that of the poly(ITC), a glass transition temperature was not found. The availability of the pendant hydroxyl group is expected to enhance the biodegradability of the polymer and serves in a variety of potential biomedical applications such as polymeric drug delivery systems.


Subject(s)
Biocompatible Materials/chemistry , Polycarboxylate Cement/chemistry , Polymers/chemistry , Tartrates/chemistry , Carbonates/chemistry , Catalysis , Chemistry/methods , Drug Delivery Systems , Enzymes, Immobilized , Fungal Proteins , Kinetics , Lipase/chemistry , Magnetic Resonance Spectroscopy , Models, Chemical , Molecular Weight , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL