Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7: 41795, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165482

ABSTRACT

The role for c-Jun N-terminal Kinase (JNK) in the control of feeding and energy balance is not well understood. Here, by use of novel and highly selective JNK inhibitors, we investigated the actions of JNK in the control of feeding and body weight homeostasis. In lean mice, intraperitoneal (i.p.) or intracerebroventricular (i.c.v.) administration of SR-3306, a brain-penetrant and selective pan-JNK (JNK1/2/3) inhibitor, reduced food intake and body weight. Moreover, i.p. and i.c.v. administrations of SR11935, a brain-penetrant and JNK2/3 isoform-selective inhibitor, exerted similar anorectic effects as SR3306, which suggests JNK2 or JNK3 mediates aspect of the anorectic effect by pan-JNK inhibition. Furthermore, daily i.p. injection of SR3306 (7 days) prevented the increases in food intake and weight gain in lean mice upon high-fat diet feeding, and this injection paradigm reduced high-fat intake and obesity in diet-induced obese (DIO) mice. In the DIO mice, JNK inhibition sensitized leptin's anorectic effect, and enhanced leptin-induced STAT3 activation in the hypothalamus. The underlying mechanisms likely involve the downregulation of SOCS3 by JNK inhibition. Collectively, our data suggest that JNK activity promotes positive energy balance, and the therapeutic intervention inhibiting JNK activities represents a promising approach to ameliorate diet-induced obesity and leptin resistance.


Subject(s)
Eating/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Leptin/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Diet, High-Fat , Energy Metabolism/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Obesity/genetics , Obesity/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Weight Loss/drug effects
3.
J Med Chem ; 59(6): 2269-300, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26486225

ABSTRACT

Rho kinases (ROCKs) belong to the serine-threonine family, the inhibition of which affects the function of many downstream substrates. As such, ROCK inhibitors have potential therapeutic applicability in a wide variety of pathological conditions including asthma, cancer, erectile dysfunction, glaucoma, insulin resistance, kidney failure, neuronal degeneration, and osteoporosis. To date, two ROCK inhibitors have been approved for clinical use in Japan (fasudil and ripasudil) and one in China (fasudil). In 1995 fasudil was approved for the treatment of cerebral vasospasm, and more recently, ripasudil was approved for the treatment of glaucoma in 2014. In this Perspective, we present a comprehensive review of the physiological and biological functions for ROCK, the properties and development of over 170 ROCK inhibitors as well as their therapeutic potential, the current status, and future considerations.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , Animals , Humans , Protein Kinase Inhibitors/chemical synthesis , Structure-Activity Relationship , rho-Associated Kinases/metabolism
4.
ACS Med Chem Lett ; 6(4): 413-8, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25893042

ABSTRACT

A novel series of 2-aminopyridopyrimidinone based JNK (c-jun N-terminal kinase) inhibitors were discovered and developed. Structure-activity relationships (SARs) were systematically developed utilizing biochemical and cell based assays and in vitro and in vivo drug metabolism and pharmacokinetic (DMPK) studies. Through the optimization of lead compound 1, several potent and selective JNK inhibitors with high oral bioavailability were developed. Inhibitor 13 was a potent JNK3 inhibitor (IC50 = 15 nM), had high selectivity against p38 (IC50 > 10 µM), had high potency in functional cell based assays, and had high stability in human liver microsome (t 1/2 = 76 min), a clean CYP-450 inhibition profile, and excellent oral bioavailability (%F = 87). Moreover, cocrystal structures of compounds 13 and 22 in JNK3 were solved at 2.0 Å. These structures elucidated the binding mode (Type-I binding) and can pave the way for further inhibitor design of this pyridopyrimidinone scaffold for JNK inhibition.

5.
Mol Cell Biol ; 35(11): 1992-2006, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25825522

ABSTRACT

Serum glucocorticoid kinase 1 (SGK1) has been shown to be protective in models of Parkinson's disease, but the details by which it confers benefit is unknown. The current study was designed to investigate the details by which SGK1 confers neuroprotection. To do this we employed a cellular neurodegeneration model to investigate c-Jun N-terminal kinase (JNK) signaling and endoplasmic reticulum (ER) stress induced by 6-hydroxydopamine. SGK1-expressing adenovirus was created and used to overexpress SGK1 in SH-SY5Y cells, and dexamethasone was used to increase endogenous expression of SGK1. Oxidative stress, mitochondrial dysfunction, and cell death were monitored to test the protective effect of SGK1. To investigate the effect of SGK1 overexpression in vivo, SGK1-expressing adenovirus was injected into the striatum of mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and protection of dopaminergic neurons was quantitatively assessed by tyrosine hydroxylase immunohistochemistry. SGK1 overexpression was found to decrease reactive oxygen species generation, alleviate mitochondrial dysfunction, and rescue cell death in vitro and in vivo by inactivating mitogen-activated protein kinase kinase 4 (MKK4), JNK, and glycogen synthase kinase 3ß (GSK3ß) and thereby decreasing ER and oxidative stress. These results suggest that therapeutic strategies for activation of SGK1 may have the potential to be neuroprotective by deactivating the JNK and GSK3ß pathways.


Subject(s)
Immediate-Early Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neurotoxins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Oxidopamine/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
6.
J Med Chem ; 58(4): 1846-61, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25621531

ABSTRACT

The discovery/optimization of bis-aryl ureas as Limk inhibitors to obtain high potency and selectivity and appropriate pharmacokinetic properties through systematic SAR studies is reported. Docking studies supported the observed SAR. Optimized Limk inhibitors had high biochemical potency (IC50 < 25 nM), excellent selectivity against ROCK and JNK kinases (>400-fold), potent inhibition of cofilin phosphorylation in A7r5, PC-3, and CEM-SS T cells (IC50 < 1 µM), and good in vitro and in vivo pharmacokinetic properties. In the profiling against a panel of 61 kinases, compound 18b at 1 µM inhibited only Limk1 and STK16 with ≥80% inhibition. Compounds 18b and 18f were highly efficient in inhibiting cell-invasion/migration in PC-3 cells. In addition, compound 18w was demonstrated to be effective on reducing intraocular pressure (IOP) on rat eyes. Taken together, these data demonstrated that we had developed a novel class of bis-aryl urea derived potent and selective Limk inhibitors.


Subject(s)
Lim Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Urea/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Lim Kinases/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Rats , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
7.
J Med Chem ; 57(23): 10013-30, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25393557

ABSTRACT

The c-jun N-terminal kinase 3 (JNK3) is expressed primarily in the brain. Numerous reports have shown that inhibition of JNK3 is a promising strategy for treatment of neurodegeneration. The optimization of aminopyrazole-based JNK3 inhibitors with improved potency, isoform selectivity, and pharmacological properties by structure-activity relationship (SAR) studies utilizing biochemical and cell-based assays, and structure-based drug design is reported. These inhibitors had high selectivity over JNK1 and p38α, minimal cytotoxicity, potent inhibition of 6-OHDA-induced mitochondrial membrane potential dissipation and ROS generation, and good drug metabolism and pharmacokinetic (DMPK) properties for iv dosing. 26n was profiled against 464 kinases and was found to be highly selective hitting only seven kinases with >80% inhibition at 10 µM. Moreover, 26n showed good solubility, good brain penetration, and good DMPK properties. Finally, the crystal structure of 26k in complex with JNK3 was solved at 1.8 Å to explore the binding mode of aminopyrazole based JNK3 inhibitors.


Subject(s)
Benzamides/chemical synthesis , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Piperidines/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Pyrazoles/chemical synthesis , Animals , Benzamides/pharmacokinetics , Benzamides/pharmacology , Crystallography, X-Ray , Drug Design , Humans , Inhibitory Concentration 50 , Mice , Microsomes, Liver/drug effects , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Oxidopamine/pharmacology , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Isoforms/antagonists & inhibitors , Pyrazoles/pharmacology , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 24(1): 161-4, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24332487

ABSTRACT

The design and synthesis of isoxazole 3 is described, a potent JNK inhibitor with two fold selectivity over p38. Optimization of this scaffold led to compounds 27 and 28 which showed greatly improved selectivity over p38 by maintaining the JNK3 potency of compound 3. Extensive SAR studies will be described as well as preliminary in vivo data of the two lead compounds.


Subject(s)
Isoxazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Dose-Response Relationship, Drug , Humans , Isoxazoles/administration & dosage , Isoxazoles/chemical synthesis , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Rats , Structure-Activity Relationship , Tissue Distribution
9.
Expert Opin Ther Pat ; 24(3): 295-307, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24283930

ABSTRACT

INTRODUCTION: The Rho kinase/ROCK is critical in vital signal transduction pathways central to many essential cellular activities. Since ROCK possess multiple substrates, modulation of ROCK activity is useful for treatment of many diseases. AREAS COVERED: Significant progress has been made in the development of ROCK inhibitors over the past two years (Jan 2012 to Aug 2013). Patent search in this review was based on FPO IP Research and Communities and Espacenet Patent Search. In this review, patent applications will be classified into four groups for discussions. The grouping is mainly based on structures or scaffolds (groups 1 and 2) and biological functions of ROCK inhibitors (groups 3 and 4). These four groups are i) ROCK inhibitors based on classical structural elements for ROCK inhibition; ii) ROCK inhibitors based on new scaffolds; iii) bis-functional ROCK inhibitors; and iv) novel applications of ROCK inhibitors. EXPERT OPINION: Although currently only one ROCK inhibitor (fasudil) is used as a drug, more drugs based on ROCK inhibition are expected to be advanced into market in the near future. Several directions should be considered for future development of ROCK inhibitors, such as soft ROCK inhibitors, bis-functional ROCK inhibitors, ROCK2 isoform-selective inhibitors, and ROCK inhibitors as antiproliferation agents.


Subject(s)
Protein Kinase Inhibitors/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , Animals , Drug Design , Humans , Legislation, Drug , Molecular Structure , Molecular Targeted Therapy , Patents as Topic , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , rho-Associated Kinases/metabolism
10.
ACS Chem Biol ; 8(8): 1747-54, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23751758

ABSTRACT

Both JNK and LRRK2 are associated with Parkinson's disease (PD). Here we report a reasonably selective and potent kinase inhibitor (compound 6) that bound to both JNK and LRRK2 (a dual inhibitor). A bidentate-binding strategy that simultaneously utilized the ATP hinge binding and a unique protein surface site outside of the ATP pocket was applied to the design and identification of this kind of inhibitor. Compound 6 was a potent JNK3 and modest LRRK2 dual inhibitor with an enzyme IC50 value of 12 nM and 99 nM (LRRK2-G2019S), respectively. Compound 6 also exhibited good cell potency, inhibited LRRK2:G2019S-induced mitochondrial dysfunction in SHSY5Y cells, and was demonstrated to be reasonably selective against a panel of 116 kinases from representative kinase families. Design of such a probe molecule may help enable testing if dual JNK and LRRK2 inhibitions have added or synergistic efficacy in protecting against neurodegeneration in PD.


Subject(s)
Benzofurans/chemistry , Enzyme Inhibitors/chemistry , Indazoles/chemistry , MAP Kinase Kinase 4/antagonists & inhibitors , Molecular Probes , Protein Serine-Threonine Kinases/antagonists & inhibitors , Benzofurans/pharmacology , Binding Sites , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Indazoles/pharmacology , Inhibitory Concentration 50 , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Models, Molecular , Molecular Probes/chemistry , Molecular Probes/pharmacology , Protein Binding/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
12.
J Med Chem ; 56(9): 3568-81, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23570561

ABSTRACT

RhoA and its downstream effector ROCK mediate stress fiber formation and cell contraction through their effects on the phosphorylation of myosin light chain (MLC). Inhibition of the RhoA/ROCK pathway has proven to be a promising strategy for several indications such as cardiovascular disease, glaucoma, and inflammatory disease. In 2010, our group reported urea-based ROCK inhibitors as potential antiglaucoma agents. These compounds showed potent IC50 values in enzymatic and cell-based assays and significant intraocular pressure (IOP)-lowering effects in rats (∼7 mmHg). (22) To develop more advanced ROCK inhibitors targeting various potential applications (such as myocardial infarction, erectile dysfunction, multiple sclerosis, etc.) in addition to glaucoma, a thorough SAR for this urea-based scaffold was studied. The detailed optimization process, counter-screening, and in vitro and in vivo DMPK studies are discussed. Potent and selective ROCK inhibitors with various in vivo pharmacokinetic properties were discovered.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Urea/chemical synthesis , Urea/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Cell Line , Chemistry Techniques, Synthetic , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Protein Conformation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Rats , Structure-Activity Relationship , Substrate Specificity , Urea/metabolism , Urea/pharmacokinetics , rho-Associated Kinases/chemistry , rho-Associated Kinases/metabolism
13.
Bioorg Med Chem Lett ; 23(6): 1592-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23416002

ABSTRACT

SAR and lead optimization studies for Rock inhibitors based on amino acid-derived quinazolines are described. Studies demonstrated that these amino acid derived quinazolinones were mainly pan-Rock (I & II) inhibitors. While selectivity against other kinases could be achieved, selectivity for most of these compounds against PKA was not achieved. This is distinct from Rock inhibitors based on non-amino acid derived quinazolinones, where high selectivity against PKA could be obtained.(22) The inhibitors presented here in some cases possessed sub-nanomolar inhibition of Rock, nanomolar potency in ppMLC cell based assays, low to fair cytochrome P-450 inhibition, and good human microsomal stability.


Subject(s)
Amino Acids/chemistry , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Quinazolines/chemistry , rho-Associated Kinases/antagonists & inhibitors , Binding Sites , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Microsomes/metabolism , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Quinazolines/chemical synthesis , Quinazolines/metabolism , Structure-Activity Relationship , rho-Associated Kinases/metabolism
14.
J Biol Chem ; 288(6): 4000-11, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23258542

ABSTRACT

To build upon recent findings that mitochondrial JNK signaling is inhibited by selectively blocking the interaction between JNK and Sab, we utilized a cell-permeable peptide to demonstrate that ischemia/reperfusion (I/R) injury could be protected in vivo and that JNK mitochondrial signaling was the mechanism by which reactive oxygen species (ROS) generation, mitochondrial dysfunction, and cardiomyocyte cell death occur. We also demonstrated that 5 mg/kg SR-3306 (a selective JNK inhibitor) was able to protect against I/R injury, reducing infarct volume by 34% (p < 0.05) while also decreasing I/R-induced increases in the activity of creatine phosphokinase and creatine kinase-MB. TUNEL staining showed that the percent TUNEL positive nuclei in rat hearts increased 10-fold after I/R injury and that this was reduced 4-fold (p < 0.01) by SR-3306. These data suggest that blocking JNK mitochondrial translocation or JNK inhibition prevents ROS increases and mitochondrial dysfunction and may be an effective treatment for I/R-induced cardiomyocyte death.


Subject(s)
MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Mitochondria, Heart/enzymology , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Animals , Cell Death , Cell Line , Creatine Kinase/genetics , Creatine Kinase/metabolism , Creatine Kinase, MB Form/genetics , Creatine Kinase, MB Form/metabolism , Humans , MAP Kinase Kinase 4/antagonists & inhibitors , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Mitochondrial Proteins/genetics , Muscle Proteins/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Protein Transport/genetics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
15.
J Biol Chem ; 288(2): 1079-87, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23184940

ABSTRACT

Because oxidative stress and mitochondrial dysfunction are well known contributors to Parkinson disease (PD), we set out to investigate the role mitochondrial JNK plays in the etiology of 6-hydroxydopamine-induced (6-OHDA) oxidative stress, mitochondrial dysfunction, and neurotoxicity in SHSY5Y cells and neuroprotection and motor behavioral protection in vivo. To do this, we utilized a cell-permeable peptide of the outer mitochondrial membrane protein, Sab (SH3BP5), as an inhibitor of JNK mitochondrial translocation. In vitro studies showed that 6-OHDA induced JNK translocation to the mitochondria and that inhibition of mitochondrial JNK signaling by Tat-Sab(KIM1) protected against 6-OHDA-induced oxidative stress, mitochondrial dysfunction, and neurotoxicity. Administration of Tat-Sab(KIM1) via an intracerebral injection into the mid-forebrain bundle increased the number of tyrosine hydroxylase immunoreactive neurons in the substantia nigra pars compacta by 2-fold (p < 0.05) in animals lesioned with 6-OHDA, compared with animals treated only with 6-OHDA into the nigrostriatal pathway. In addition, Tat-Sab(KIM1) decreased the d-amphetamine-induced unilateral rotations associated with the lesion by 30% (p < 0.05). Steady-state brain levels of Tat-Sab(KIM1) at day 7 were 750 nm, which was ∼3.4-fold higher than the IC(50) for this peptide versus Sab protein. Collectively, these data suggest that 6-OHDA induced JNK translocation to the mitochondria and that blocking this translocation reduced oxidative stress, mitochondrial dysfunction, and neurotoxicity both in vitro and in vivo. Moreover, the data suggest that inhibitors that block association of JNKs with the mitochondria may be useful neuroprotective agents for the treatment of Parkinson disease.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Oxidopamine/toxicity , Animals , Blotting, Western , Cell Death , Cell Line , Dopamine/metabolism , Gene Silencing , Humans , Immunohistochemistry , In Vitro Techniques , Mitochondria/metabolism , Neurons/cytology , Protein Transport , Rats , Rats, Sprague-Dawley , Signal Transduction
16.
Structure ; 20(12): 2174-84, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23142346

ABSTRACT

c-Jun N-terminal (JNK) family kinases have a common peptide-docking site used by upstream activating kinases, substrates, scaffold proteins, and phosphatases, where the ensemble of bound proteins determines signaling output. Although there are many JNK structures, little is known about mechanisms of allosteric regulation between the catalytic and peptide-binding sites, and the activation loop, whose phosphorylation is required for catalytic activity. Here, we compare three structures of unliganded JNK3 bound to different peptides. These were compared as a class to structures that differ in binding of peptide, small molecule ligand, or conformation of the kinase activation loop. Peptide binding induced an inhibitory interlobe conformer that was reversed by alterations in the activation loop. Structure class analysis revealed the subtle structural mechanisms for allosteric signaling between the peptide-binding site and activation loop. Biochemical data from isothermal calorimetry, fluorescence energy transfer, and enzyme inhibition demonstrated affinity differences among the three peptides that were consistent with structural observations.


Subject(s)
Mitogen-Activated Protein Kinase 10/chemistry , Activating Transcription Factor 2/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Allosteric Regulation , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/chemistry , Models, Molecular , Oligopeptides/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Substrate Specificity
17.
Stem Cells ; 30(5): 975-87, 2012 May.
Article in English | MEDLINE | ID: mdl-22367737

ABSTRACT

Large scale expansion of human mesenchymal stem cells (MSCs) is routinely performed for clinical therapy. In contrast, developing protocols for large scale expansion of primary mouse MSCs has been more difficult due to unique aspects of rodent biology. Currently, established methods to isolate mouse MSCs select for rapidly dividing subpopulations that emerge from bone marrow cultures following long-term (months) expansion in atmospheric oxygen. Herein, we demonstrate that exposure to atmospheric oxygen rapidly induced p53, TOP2A, and BCL2-associated X protein (BAX) expression and mitochondrial reactive oxygen species (ROS) generation in primary mouse MSCs resulting in oxidative stress, reduced cell viability, and inhibition of cell proliferation. Alternatively, procurement and culture in 5% oxygen supported more prolific expansion of the CD45(-ve) /CD44(+ve) cell fraction in marrow, produced increased MSC yields following immunodepletion, and supported sustained MSC growth resulting in a 2,300-fold increase in cumulative cell yield by fourth passage. MSCs cultured in 5% oxygen also exhibited enhanced trilineage differentiation. The oxygen-induced stress response was dependent upon p53 since siRNA-mediated knockdown of p53 in wild-type cells or exposure of p53(-/-) MSCs to atmospheric oxygen failed to induce ROS generation, reduce viability, or arrest cell growth. These data indicate that long-term culture expansion of mouse MSCs in atmospheric oxygen selects for clones with absent or impaired p53 function, which allows cells to escape oxygen-induced growth inhibition. In contrast, expansion in 5% oxygen generates large numbers of primary mouse MSCs that retain sensitivity to atmospheric oxygen, and therefore a functional p53 protein, even after long-term expansion in vitro.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mesenchymal Stem Cells/metabolism , Oxygen/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Bone Marrow Cells/cytology , Cells, Cultured , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Poly-ADP-Ribose Binding Proteins , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
J Biol Chem ; 287(16): 13291-302, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22351776

ABSTRACT

c-Jun N-terminal kinase (JNK) is a stress signal transducer linked to cell death, and survival. JNK1 has been implicated in obesity, glucose intolerance, and insulin resistance. In this study we report the kinetic mechanism for JNK1ß1 with transcription factors ATF2 and c-Jun along with interaction kinetics for these substrates. JNK1ß1 followed a random sequential mechanism forming a ternary complex between JNK-substrate-ATP. K(m) for ATF2 and c-Jun was 1.1 and 2.8 µM, respectively. Inhibition studies using adenosine 5'-(ß,γ-methylenetriphosphate) and a peptide derived from JNK interacting protein 1 (JIP1) supported the proposed kinetic mechanism. Biolayer interferometry studies showed that unphosphorylated JNK1ß1 bound to ATF2 with similar affinity as it did to c-Jun (K(D) = 2.60 ± 0.34 versus 1.00 ± 0.35 µM, respectively). The presence of ATP increased the affinity of unphosphorylated JNK1ß1 for ATF2 and c-Jun, to 0.80 ± 0.04 versus 0.65 ± 0.07 µM, respectively. Phosphorylation of JNK1ß1 decreased the affinity of the kinase for ATF2 to 11.0 ± 1.1 µM and for c-Jun to 17.0 ± 7.5 µM in the absence of ATP. The presence of ATP caused a shift in the K(D) of the active kinase for ATF2 to 1.70 ± 0.25 µM and for c-Jun of 3.50 ± 0.95 µM. These results are the first kinetic and biochemical characterization of JNK1ß1 and uncover some of the differences in the enzymatic activity of JNK1ß1 compared with other variants and suggest that ATP binding or JNK phosphorylation could induce changes in the interactions with substrates, activators, and regulatory proteins.


Subject(s)
Activating Transcription Factor 2/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Alternative Splicing/physiology , Cell Death/physiology , Enzyme Activation/physiology , Escherichia coli/genetics , Gene Expression Regulation, Enzymologic/physiology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Kinetics , Stress, Physiological/physiology , Substrate Specificity/physiology
19.
Chem Biol ; 19(1): 140-54, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22284361

ABSTRACT

The mitogen-activated kinases JNK1/2/3 are key enzymes in signaling modules that transduce and integrate extracellular stimuli into coordinated cellular response. Here, we report the discovery of irreversible inhibitors of JNK1/2/3. We describe two JNK3 cocrystal structures at 2.60 and 2.97 Å resolution that show the compounds form covalent bonds with a conserved cysteine residue. JNK-IN-8 is a selective JNK inhibitor that inhibits phosphorylation of c-Jun, a direct substrate of JNK, in cells exposed to submicromolar drug in a manner that depends on covalent modification of the conserved cysteine residue. Extensive biochemical, cellular, and pathway-based profiling establish the selectivity of JNK-IN-8 for JNK and suggests that the compound will be broadly useful as a pharmacological probe of JNK-dependent signal transduction. Potential lead compounds have also been identified for kinases, including IRAK1, PIK3C3, PIP4K2C, and PIP5K3.


Subject(s)
JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary
20.
Org Lett ; 13(19): 5108-11, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21866939

ABSTRACT

A convergent synthesis of benzoquinone ansamycin analogs is described that proceeds by a sequence of metallacycle-mediated alkyne-alkyne coupling, followed by site- and stereoselective dihydroxylation and global carbamate formation. These studies have led to (1) validation of alkyne-alkyne coupling to produce geldanamycin analogs that lack the problematic quinone, (2) the discovery that C6-C7 bis-carbamate functionality is compatible with Hsp90 inhibition, and (3) the identification of 1 as a nonquinone geldanamycin-inspired paralog-selective Hsp90 inhibitor.


Subject(s)
Benzoquinones/chemistry , Biological Products/chemical synthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/chemical synthesis , Biological Products/pharmacology , Lactams, Macrocyclic/pharmacology , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...