US20030207849A1 - P38MAPK inhibitor and uses thereof - Google Patents

P38MAPK inhibitor and uses thereof Download PDF

Info

Publication number
US20030207849A1
US20030207849A1 US10/408,968 US40896803A US2003207849A1 US 20030207849 A1 US20030207849 A1 US 20030207849A1 US 40896803 A US40896803 A US 40896803A US 2003207849 A1 US2003207849 A1 US 2003207849A1
Authority
US
United States
Prior art keywords
mapk
ldl receptor
receptor expression
cell
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/408,968
Inventor
Kamal Mehta
Rajesh Singh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Arkansas
Original Assignee
University of Arkansas
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Arkansas filed Critical University of Arkansas
Priority to US10/408,968 priority Critical patent/US20030207849A1/en
Publication of US20030207849A1 publication Critical patent/US20030207849A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF ARKANSAS MED SCIS LTL ROCK
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole

Definitions

  • the present invention relates generally to the fields of molecular biology and protein biochemistry. More specifically, the present invention relates to p38 MAPK inhibitor(s) and applications of p38 MAPK inhibitor(s) in the treatment of hypercholesterolemia.
  • Mitogen-activated protein kinases are proline-directed serine-threonine-protein kinases that have important functions as mediators of cellular responses to a variety of extracellular stimuli (1-4).
  • Three subgroups of the mitogen-activated protein kinase super-family have been clearly identified: the extracellularly responsive kinases (p42/44 MAPK or ERK-1/2), the c-Jun N-terminal kinases (p46/54 JNK ) which are also known as the stress-activated protein kinases (SAPK), and the p38 MAPK (also known as RK, Mxi-2, CSBP1/2 or HOG-1-related kinases).
  • mitogen-activated protein kinase families are structurally related, they are generally activated by distinct extracellular stimuli through distinct upstream dual specificity kinases, thus comprising a series of separate mitogen-activated protein kinase cascades (5-7).
  • the best known pathway, Raf/MAPK/ERK kinase-1/2 (MEK-1/2)/p42/44 MAPK is typically strongly stimulated by growth factors and mitogenic stimuli, usually by means of a Ras-Raf-1-dependent cascade (8, 9).
  • p38 MAPK and p46/54 JNK are primarily activated by cellular stresses, including heat and osmotic shock, UV irradiation, proinflammatory cytokines, and hypoxia/reoxygenation (10-16).
  • Ten isoforms of p46/54 JNK and four isoforms of p38 MAPK have been identified in mammalian cells (17-26). No physiological role has been associated with the difference in substrate affinity of the JNKs or p38 MAPK .
  • Dual specificity kinases that activate p46/54 JNK are MAPK kinase 4 (MKK4/SEK-1) and MKK7, whereas MKK3 and MKK6 have been identified as activators of p38 MAPK , displaying some degree of selectivity for individual p38 MAPK isoforms (26-32).
  • MKK6 functions as an activating kinase for all known p38 MAPK isoforms, whereas MKK3 predominantly activates the isoform p38 MAPK ⁇ .
  • the identified substrates of mitogen-activated protein kinases are a variety of transcription factors that become activated upon their phosphorylation (1, 4, 6, 33).
  • the flavone compound PD98059 (2-(2′-amino-3′-methoxyphenyl) oxanaphthalen-4-one) is a specific inhibitor of the mammalian MEK-1/2 and has been used extensively for investigating physiological function of p42/44 MAPK pathway (34).
  • pyridinylimidazole compounds SB203580 (4-(4-Fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole) and SB202190 (4(4-Fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl) 1H-imidazole) are specific inhibitors for p38 MAPK since they selectively inhibit both p38 MAPK ⁇ - and ⁇ -isoforms, but not the ⁇ - and ⁇ -isoforms.
  • the prior art is deficient in the lack of understanding the role of p38 MAPK in the regulation of the LDL receptor expression. Further, the prior art is deficient in the lack of effective means of applying p38 MAPK inhibitors to treat hypercholesterolemia.
  • the present invention fulfills this long-standing need and desire in the art.
  • the present invention addresses directly the physiological role of p38 MAPK in the regulation of LDL receptor expression by using highly specific pharmacological and molecular tools.
  • Results presented demonstrate that simple inhibition of the p38 MAPK basal activity is sufficient to induce LDL receptor expression.
  • Co-transfection studies established that SB202190-induced LDL receptor expression is mediated by the activation of p42/44 MAPK resulting from the inhibition of p38 MAPK ⁇ -isoform. Therefore, in intact cells, p38 MAPK negatively regulates the p42/44 MAPK and the responses mediated by this kinase. It is speculated that cross-talk between these mitogen-activated protein kinases, which mediate the effects of numerous extracellular stimuli, could be crucial for controlling a wide array of biological processes.
  • a compound for inducing low density lipoprotein receptor expression wherein the compound is a p38 MAPK inhibitor or a compound that activates p42/44 MAPK .
  • the representative examples of such compounds are SB202190 and SB203580.
  • a method of inducing LDL receptor expression in a cell by administering to the cell a compound disclosed herein.
  • the cell is either hepatic or nonhepatic.
  • the compound induces the LDL receptor expression by 6-8 fold and further reduces cholesterol level in the cell.
  • a method of treating an individual having hypercholesterolemia by administering a compound disclosed herein.
  • the compound is administered at a concentration range of from about 1 ⁇ M to about 100 ⁇ M.
  • FIG. 1 shows selective induction of LDL receptor expression by SB202190. 5 ⁇ 10 5 cells were plated on day 0. On day 2, cells were refed with fresh medium. On day 4, cells were either left untreated (0) or treated for the indicated times with SB202190 (2.5 ⁇ M) in medium containing 0.5% FBS. Total RNA was isolated and subjected to Northern blotting to determine amounts of LDL receptor, squalene synthase (SS) genes, and actin mRNAs. Autoradiographs were quantitated densitometrically. LDL receptor mRNA levels were normalized to actin mRNA levels. In the bottom panel, results are expressed as the fold induction by SB 202190 as compared with uninduced cells (set at 1). Values obtained are the averages of three separate experiments, with the standard deviations shown.
  • SS squalene synthase
  • FIG. 2 shows that SB202190 induces LDL receptor expression in a sterol-sensitive manner irrespective of concentrations.
  • HepG2 cells were grown as described in FIG. 1 On day 4, medium was changed to either 10% LPDS alone or 10% LPDS supplemented with 25-hydroxycholesterol (2 ⁇ g/ml) and cholesterol (10 ⁇ g/ml), and after 2 hours, cells were treated with indicated concentrations of SB202190 for an additional 4 hours.
  • Levels of LDL receptor and SS mRNAs were determined by Northern analysis and densitometry as described previously. RNA levels were normalized by comparison with levels of actin. Values obtained from cells cultured in the absence of sterols and SB202190 were set at 1. Values shown are the average of two different experiments. The experiment was repeated two times with similar results.
  • FIG. 3 shows activation of p38 MAPK ⁇ -isoform interferes with LDL receptor promoter activity.
  • HepG2 cells were co-transfected with LDL receptor-luciferase reporter (plasmid A) (46) and expression vectors encoding MKK6b, MKK6b(A), MKK6b(E), p38 MAPK ⁇ -isoform, p38 MAPK ⁇ -isoform, p38 MAPK ⁇ -isoform (AF) as indicated. Following transfection, cells were washed three times with PBS and maintained in 0.5% FBS. After 16 hours, cells were harvested and luciferase activity was determined and normalized to the protein content of each extract.
  • Luciferase activity expressed by cells transfected with empty vector was given an arbitrary value of 1.
  • the results are presented as means ⁇ standard errors and represent at least four individual experiments.
  • the amount of DNA used were: plasmid A (0.75 ⁇ g/well), MKK6b, MKK6b(A), MKK6b(E) (0.6 ⁇ g/well), p38 MAPK (x-isoform, cc(AF) mutant, p38 MAPK ⁇ -isoform (0.2 ⁇ g/well).
  • FIG. 4 shows SB202190-induced LDL receptor expression is mediated by p42/44 MAPK signaling cascade.
  • FIG. 4A shows kinetics of p42/44 MAPK , and MEK-1/2 activation by SB202190. 2 ⁇ 10 5 cells were grown and treated as described in FIG. 1. After the indicated times, equal amounts of cell lysates were blotted with anti-phospho-p42/44 MAPK antibody, phosphorylation-independent p42/44 MAPK antibody, anti-phospho p46/54 JNK antibody, anti-phospho-MEK-1/2, or phosphorylation-independent MKP-1.
  • FIG. 4B shows that HepG2 cells were grown as described in FIG. 1, and were either untreated or treated with SB202190 (2.5 ⁇ M) for 4 h either in the absence or presence of indicated concentrations of PD98059 that was added 30 min prior to SB202190 (2.5 ⁇ M) addition.
  • Total RNA was subjected to Northern blot analysis, and the filter was hybridized with a 32 P-labeled LDL receptor probe. Values shown are the averages of two different experiments. Ethidium bromide staining of RNA gel before blotting onto a nitrocellulose to demonstrate equal loading of RNA in all lanes. Values obtained from the control cells grown in the absence of SB202190 or PD98059 were arbitrarily set at 1.
  • FIG. 4C shows that cells were grown as described in FIG. 2.
  • medium was changed to either 10% LPDS alone or 10% LPDS supplemented with three different concentrations of 25-hydroxycholesterol (2 ⁇ g/ml, 5 ⁇ g/ml, or 10 ⁇ g/ml) together with cholesterol (10 ⁇ g/ml).
  • 25-hydroxycholesterol 2 ⁇ g/ml, 5 ⁇ g/ml, or 10 ⁇ g/ml
  • cholesterol 10 ⁇ g/ml
  • FIG. 5 shows that SB202190 induces LDL receptor expression and phosphorylation of p42/44 MAPK in HeLa cells, and pretreatment with PD98059 inhibited SB20219-induced LDL receptor expression.
  • FIG. 5A shows that 1 ⁇ 10 5 cells were plated on day 0, and were refed with fresh medium on day 2. On day 4, cells were treated with SB202190 (2.5 ⁇ g/ml) in 0.5% FBS. After the indicated times, cells were lysed in SDS sample buffer, and subjected to Western blotting with anti-phospho-p42/44 MAPK or phosphorylation-independent anti-p42/44 MAPK . Equal amounts of p38 MAPK was observed in all the lanes (data not shown).
  • FIG. 5B shows that HeLa cells were either untreated or treated with SB202190 (2.5 ⁇ M) for 4 h either in the absence or presence of of PD98059 (5 ⁇ M). PD98059 was added 30 min prior to addition of SB202190. After the indicated times, total RNA was isolated and subjected to Northern blotting. RNA gel stained with ethidium bromide before blotting onto nitrocellulse is shown to demonstrate equal loading of RNA.
  • FIG. 6 shows modulation of p42/44 MAPK activity does not affect p38 MAPK phosphorylation in response to a variety of stress-inducers in HepG2- ⁇ Raf-1:ER cells.
  • Cells were either left untreated or stimulated with 1 ⁇ M ICI 182,780 for 1.5 h, and then treated with either anisomycin (50 ng/ml) or IL-1 ⁇ (5 ng/ml) for 30 min. Cells were then lysed in SDS sample buffer, and equal amounts of whole cell lysates were separated by SDS-PAGE in 10% gels.
  • Activated MAPKs levels were analyzed by blotting using anti-phospho-p42/44 MAPK , and anti-phospho-p38 MAPK . Protein levels were analyzed by using phosphorylation-independent anti-p42/44 MAPK following electrotransfer of total proteins onto nitrocellulose. Similar results were obtained in three separate experiments.
  • FIG. 7 shows that SB202190-induced LDL receptor expression does not require new protein synthesis.
  • HepG2 cells were grown as described in FIG. 1. On day 4, cells cultured for 1 h in 0.5% FBS containing different amounts of cycloheximide (1 ⁇ g/ml, 2 ⁇ g/ml, and 5 ⁇ g/ml) were then incubated with SB202190 (2.5 ⁇ M) for an additional 4 h. LDL receptor and actin mRNA levels were analyzed by Northern blotting of total RNA. Values obtained from cells with no additions were arbitrarily set at 1. The experiment was repeated two times with similar results. Results shown are representative of one experiment.
  • FIG. 8 shows effects of different protein kinase C inhibitors on SB202190-induced LDL receptor expression in HepG2 cells.
  • FIG. 8A HepG2 cells were pretreated for 30 minutes with the indicated concentration of the above inhibitors and then treated with 2.5 ⁇ M SB202190 for an additional 4 hours.
  • Total RNA was subjected to Northern blotting for measurement of LDL receptor mRNA levels. Ethidium bromide staining of RNA gel before blotting onto a nitrocellulose paper to demonstrate equal loading of RNA in all lanes. Similar results were obtained in four seaparate experiments.
  • FIG. 8B shows that staurosporine blocked SB202190-induced p42/44 MAPK and MEK-1/2 activation.
  • HepG2 cells were cultured in the absence or presence of 1 ⁇ M staurosporine or 5 ⁇ M PD98059 for 30 min.
  • SB202190 2.5 ⁇ M was then added to the cells for 4 h, cell lysate was subjected to immunoblotting to measure the phosphorylation levels of p42/44 MAPK and MEK-1/2.
  • FIG. 9 shows a suggested model for cross-talk between p38 MAPK and p42/44 MAPK signaling pathways for regulating LDL receptor expression.
  • the present invention examines the role of p38 MAPK in the regulation of the LDL receptor expression and demonstrates that there is a cross-talk between p42/44 MAPK and p38 MAPK signaling cascades, and p38 MAPK negatively regulates LDL receptor expression via the p42/44 MAPK signaling cascade.
  • the present invention reports that SB202190 alone, a specific inhibitor of p38 MAPK , induces low density lipoprotein (LDL) receptor expression (6-8 fold) in a sterol-sensitive manner in HepG2 cells. Consistent with this finding, selective activation of the p38 MAPK signaling pathway by expression of MKK6b (E), a constitutive activator of p38 MAPK , significantly reduced LDL receptor promoter activity. Expression of the p38 MAPK ⁇ -isoform had a similar effect, whereas, expression of the p38 MAPK ⁇ II-isoform had no effect on LDL receptor promoter activity.
  • LDL low density lipoprotein
  • a compound for inducing low density lipoprotein receptor expression wherein the compound is a p38 MAPK inhibitor or a compound that activates p42/44 MAPK .
  • the representative examples of such compounds are SB202190 and SB203580.
  • a method of inducing LDL receptor expression in a cell by administering to the cell with the compound disclosed herein.
  • the cell is either hepatic or nonhepatic.
  • the compound induces the LDL receptor expression by 6-8 fold and further reduces cholesterol level in the cell.
  • a method of treating an individual having hypercholesterolemia by administering the compound disclosed herein.
  • the compound is administered at a concentration range of from about 1 ⁇ M to about 100 ⁇ M.
  • Cycloheximide, calphostin C, PD98059, and SB202190 were purchased from Calbiochem (San Diego, Calif.).
  • Phospho-specific antibodies to the activated forms of p42/44 MAPK (Thr 202/Tyr 204), p46/54 JNK (Thr 183/Tyr 188), p38 MAPK (Thr 180/Tyr 182), and MEK-1/2 (Ser 217/221) were purchased from New England Biolabs (Beverly, Mass.).
  • Antibodies to p42/44 MAPK , mitogen-activated protein kinase phosphatase-1 (MKP-1) were obtained from Santa Cruz Biotechnology (Santa Cruz, Calif.).
  • IL-1 ⁇ and TNF were purchased from R & D Systems Inc. (Minneapolis, Minn.). TRIzol and all tissue culture supplies were from Life Technologies Inc. (Gaithersburg, Md.). Zeta probe blotting membrane and the protein assay reagent were purchased from Bio-Rad (Hercules, Calif.). [ ⁇ - 32 P]dCTP (3000 Ci/mmol) was obtained from Dupont (Boston, Mass.), and the enhanced chemiluminescence (ECL) detection kit was obtained from Amersham International (Arlington Heights, Ill.).
  • pSV- ⁇ -galactosidase (pSV- ⁇ -Gal) vector was purchased from Promega, and was used as a positive control for monitoring transfection efficiencies of HepG2 cells (24).
  • Dual-Light chemiluminescent reporter gene assay system for the combined detection of luciferase and ⁇ -galactosidase was purchased from TROPIX, Inc.
  • Human hepatoma cell line HepG2 and its derivative HepG2- ⁇ Raf-1:ER cell line that stably expresses the ⁇ Raf-1:ER chimera were maintained as monolayer cultures in a humidified 5% CO 2 atmosphere at 37° C. in Eagle's minimum essential medium (EMEM) (BioWhittaker, MD) supplemented with 10% fetal bovine serum (FBS) (Life Technologies Inc.), 2 mM L-glutamine, 20 units/ml penicillin and 20 ⁇ g/ml streptomycin sulphate.
  • EMEM Eagle's minimum essential medium
  • FBS fetal bovine serum
  • Proteins were fractionated by SDS-PAGE with an 10% acrylamide separation gel, and the separated proteins were transferred to nitrocellulose, and processed as described previously (43, 44). Briefly, membranes were incubated in 20 mM Tris-HCl, pH 7.6, 137 mM NaCl, 0.2% v/v Tween-20 (Tris/NaCl/Tween-20) with 5% w/v non-fat dried milk for 1 h, washed in Tris/NaCl/Tween-20 (3 ⁇ 5 min), and incubated for 1 h with primary antibody in Tris/NaCl/Tween-20 containing 1% milk at room temperature for non phospho-specific antibodies and overnight at 4° C. for phospho-specific antibodies.
  • Tris/NaCl/Tween-20 Tris/NaCl/Tween-20 containing 1% milk at room temperature for non phospho-specific antibodies and overnight at 4° C. for phospho-specific antibodies.
  • LDL receptor mRNA was normalized to squalene synthase (SS) (included in this study) or to ⁇ -actin (data not shown) mRNA level, and data for each point were plotted as the percentage of LDL receptor mRNA as compared to controls.
  • SS squalene synthase
  • ⁇ -actin data not shown
  • HepG2 cells were transfected by the Lipofectamine method.
  • LDL receptor promoter-luciferase expression assays HepG2 cells were seeded at a density of 1 ⁇ 10 6 cells per 6 well plates and co-transfected with 0.25 ⁇ g of relevant expression vector, or the corresponding empty vector (45, 46). Luciferase activity was measured 12-18 h after transfection. Normalization was achieved by co-transfecting 0.1 ⁇ g of pSV- ⁇ -Gal, a ⁇ -galactosidase reporter construct as an internal control for the transfection efficiency. Luciferase and P-galactosidase activities were measured according to the TROPIX protocol. Data are representive for at least three independent experiments performed in duplicate and are expressed as “fold increase in luciferase activity,” which was calculated relative to the basal level of LDL receptor promoter reporter activity (set to 1 unit) and corrected for empty vector effects for each expression vector.
  • p38 MAPK signaling pathway was used to modulate either positively or negatively the endogenous p38 MAPK activity.
  • LDL receptor transcription was monitored by transfecting HepG2 cells with a previously cloned fragment of the human LDL receptor promoter fused to the luciferase reporter gene (46), together with the relevant constructs.
  • p38 MAPK activity can be stimulated by coexpression of p38 MAPK with MKK6b, or by expression of constitutively activated MKK6b(E), in which the activating phosphorylation residues Ser207 and Thr211 were replaced by glutamic acids (19, 50).
  • MKK6b(E) reduced luciferase gene expression many fold, when compared with the luciferase expression in control cells transfected with the empty vector (FIG. 3). This dramatic down-regulation of the LDL receptor reporter expression in cells overexpressing the constitutively active MKK6b mutant most likely resulted from a decrease in p42/44 MAPK activity in these cells, as assessed by measuring activity of the co-transfected p42/44 MAPK plasmid (FIG. 3). Conversely, expression of the dominant-negative regulatory MKK6b(A) construct slightly increased LDL receptor-luciferase expression, correlating with a partial inhibition of endogenous MKK6b activity in cells expressing this mutated form of MKK6b.
  • MKK6b(E) may suppress LDL receptor promoter activity in a p38 MAPK -independent manner
  • MKK6b(E)-inhibited LDL receptor expression was due to activation of p38 MAPK .
  • HepG2 cells were transfected with the human LDL receptor promoter-luciferase reporter gene along with expression vectors encoding p38 MAPK ⁇ or ⁇ ( ⁇ II)-isoform, MKK6b, or empty vector. It is interesting to note that expression of p38 MAPK ⁇ -isoform or with MKK6b alone was able to suppress reporter gene expression mildly.
  • HepG2 ⁇ Raf:ER HepG2 ⁇ Raf:ER expressing an estradiol-dependent human Raf-1 protein kinase was generated.
  • the ⁇ Raf-1:ER chimera is activated in response to estradiol or anti-estrogen ICI 182780, thereby activating MEK-1/2 and then p42/44 MAPK (51, 52).
  • PKC Protein kinase C
  • phorbol-ester a well-known activator of protein kinase C
  • inhibitors of protein kinase C prevent agonist-induced p42/44 MAPK activation in several types of cells (44).
  • SB202190-induced p42/44 MAPK This delay in SB202190-induced p42/44 MAPK is most likely due to the late appearance of an immediate or downstream substrate of the p38 MAPK pathway either through de novo synthesis, post-translational modification or localization.
  • the lack of effect of sterols on SB202190-induced p42/44 MAPK activation supports the notion that the sterol-sensitive step either lie downstream of p42/44 MAPK or is a part of an independent signaling pathway.
  • the four isoforms are similar in size, show about 60 to 75% sequence homology, and are all activated by TNF, IL-1, ultraviolet radiation, and hyperosmolar medium. Some isoforms show a pronounced preference in tissue expression and selective interaction with upstream kinases and downstream substrates, pointing to highly specialized functions. These isoforms also differ in susceptibility to inhibition by SB202190; both ⁇ - and ⁇ -isoforms of p38 MAPK are inhibited by SB202190, whereas the ⁇ and ⁇ isoforms are insensitive (26). Therefore, it seems less likely that either the ⁇ or the ⁇ isoform is responsible for the induction of LDL receptor expression by SB202190.
  • TNF promotes a significant induction of p46/54 JNK and p38 MAPK but does not invariably induce apoptosis through induction of caspases (59).
  • concomitant inactivation of survival signals may be a prerequisite for p46/54 JNK and p38 MAPK to induce cell death (60).
  • deprivation of neurotrophic factors in PC-12 cells or ultraviolet-irradiation of NIH-3T3 cells not only activates the stress kinase cascades but also leads to a dramatic inhibition of the p42/44 MAPK pathway (56, 61).
  • the signaling cascade leading to p42/44 MAPK activation is still not known and might involve a complex combinatorial, spatial cross-talk of already synthesized messenger molecules.
  • the interplay between these signaling cascades should be an important process and dynamic balance may be critical for determining the outcome of a wide array of biological processes. Elucidation of the signaling components involved in this communication will significantly advance the ability to design novel strategies for the treatment of hypercholesterolemia and for understanding other important pathophysiological processes.

Abstract

The present invention demonstrates that p38MAPK inhibitor induces low density lipoprotein receptor expression 6-8 fold, and further provides the application of such inhibitor in the treatment of hypercholesterolemia.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This non-provisional patent application claims benefit of provisional patent application U.S. Serial No. 60/127,343, filed Apr. 1, 1999, now abandoned. [0001]
  • FEDERAL FUNDING NOTICE
  • [0002] The present invention was funded in part by National Institute of Health (Grant HL-51592-04). Consequently, the United States government has certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention [0003]
  • The present invention relates generally to the fields of molecular biology and protein biochemistry. More specifically, the present invention relates to p38[0004] MAPK inhibitor(s) and applications of p38MAPK inhibitor(s) in the treatment of hypercholesterolemia.
  • 2. Description of the Related Art [0005]
  • Mitogen-activated protein kinases (MAPKs) are proline-directed serine-threonine-protein kinases that have important functions as mediators of cellular responses to a variety of extracellular stimuli (1-4). Three subgroups of the mitogen-activated protein kinase super-family have been clearly identified: the extracellularly responsive kinases (p42/44[0006] MAPK or ERK-1/2), the c-Jun N-terminal kinases (p46/54JNK) which are also known as the stress-activated protein kinases (SAPK), and the p38MAPK (also known as RK, Mxi-2, CSBP1/2 or HOG-1-related kinases).
  • Although the mitogen-activated protein kinase families are structurally related, they are generally activated by distinct extracellular stimuli through distinct upstream dual specificity kinases, thus comprising a series of separate mitogen-activated protein kinase cascades (5-7). The best known pathway, Raf/MAPK/ERK kinase-1/2 (MEK-1/2)/p42/44[0007] MAPK is typically strongly stimulated by growth factors and mitogenic stimuli, usually by means of a Ras-Raf-1-dependent cascade (8, 9). In contrast, the other two pathways, p38MAPK and p46/54JNK are primarily activated by cellular stresses, including heat and osmotic shock, UV irradiation, proinflammatory cytokines, and hypoxia/reoxygenation (10-16). Ten isoforms of p46/54JNK and four isoforms of p38MAPK have been identified in mammalian cells (17-26). No physiological role has been associated with the difference in substrate affinity of the JNKs or p38MAPK. Dual specificity kinases that activate p46/54JNK are MAPK kinase 4 (MKK4/SEK-1) and MKK7, whereas MKK3 and MKK6 have been identified as activators of p38MAPK, displaying some degree of selectivity for individual p38MAPK isoforms (26-32). MKK6 functions as an activating kinase for all known p38MAPK isoforms, whereas MKK3 predominantly activates the isoform p38MAPK δ. Among the identified substrates of mitogen-activated protein kinases are a variety of transcription factors that become activated upon their phosphorylation (1, 4, 6, 33).
  • Since specific inhibitors of the p42/44[0008] MAPK and p38MAPK cascades were first described, they have been widely used to investigate their involvement in intracellular signal transduction pathways. The flavone compound PD98059 (2-(2′-amino-3′-methoxyphenyl) oxanaphthalen-4-one) is a specific inhibitor of the mammalian MEK-1/2 and has been used extensively for investigating physiological function of p42/44MAPK pathway (34). The pyridinylimidazole compounds SB203580 (4-(4-Fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole) and SB202190 (4(4-Fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl) 1H-imidazole) are specific inhibitors for p38MAPK since they selectively inhibit both p38MAPK α- and β-isoforms, but not the γ- and δ-isoforms. In addition, they exhibit no significant effect upon other related kinases, including other members of the mitogen-activated protein kinase families and their upstream activators (17, 35). A role for the p38MAPK has previously been identified using these inhibitors in diverse cellular processes such as lipopolysaccharide- and tumor necrosis factor-α (TNF)-induced cytokine production (17, 36), ultra-violet- and anisomycin-induced c-jun and c-fos expression (37), interleukin (IL)-2 and IL-7-mediated T-cell proliferation (38), glutamate-(39) and B cell Ag receptor-induced apoptosis (40), fibroblast growth factor-, arsenite and UVC-mediated CREB/ATF-1 phosphorylation (41, 42).
  • Analysis of the signal transduction pathways using the above inhibitors revealed a critical role for p42/44[0009] MAPK activation in induction of LDL receptor gene expression by a variety of extracellular stimuli (43-45).
  • The prior art is deficient in the lack of understanding the role of p38[0010] MAPK in the regulation of the LDL receptor expression. Further, the prior art is deficient in the lack of effective means of applying p38MAPK inhibitors to treat hypercholesterolemia. The present invention fulfills this long-standing need and desire in the art.
  • SUMMARY OF THE INVENTION
  • The present invention addresses directly the physiological role of p38[0011] MAPK in the regulation of LDL receptor expression by using highly specific pharmacological and molecular tools. Results presented demonstrate that simple inhibition of the p38MAPK basal activity is sufficient to induce LDL receptor expression. Co-transfection studies established that SB202190-induced LDL receptor expression is mediated by the activation of p42/44MAPK resulting from the inhibition of p38MAPK α-isoform. Therefore, in intact cells, p38MAPK negatively regulates the p42/44MAPK and the responses mediated by this kinase. It is speculated that cross-talk between these mitogen-activated protein kinases, which mediate the effects of numerous extracellular stimuli, could be crucial for controlling a wide array of biological processes.
  • In one embodiment of the present invention, there is provided a compound for inducing low density lipoprotein receptor expression, wherein the compound is a p38[0012] MAPK inhibitor or a compound that activates p42/44MAPK. The representative examples of such compounds are SB202190 and SB203580.
  • In another embodiment of the present invention, there is provided a method of inducing LDL receptor expression in a cell by administering to the cell a compound disclosed herein. The cell is either hepatic or nonhepatic. Preferably, the compound induces the LDL receptor expression by 6-8 fold and further reduces cholesterol level in the cell. [0013]
  • In still another embodiment of the present invention, there is provided a method of treating an individual having hypercholesterolemia by administering a compound disclosed herein. Preferably, the compound is administered at a concentration range of from about 1 μM to about 100 μM. [0014]
  • Other and further aspects, features, and advantages of the present invention will be apparent from the following description of the presently preferred embodiments of the invention given for the purpose of disclosure.[0015]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • So that the matter in which the above-recited features, advantages and objects of the invention, as well as others which will become clear, are attained and can be understood in detail, more particular descriptions of the invention briefly summarized above may be had by reference to certain embodiments thereof which are illustrated in the appended drawings. These drawings form a part of the specification. It is to be noted, however, that the appended drawings illustrate preferred embodiments of the invention and therefore are not to be considered limiting in their scope. [0016]
  • FIG. 1 shows selective induction of LDL receptor expression by SB202190. 5×10[0017] 5 cells were plated on day 0. On day 2, cells were refed with fresh medium. On day 4, cells were either left untreated (0) or treated for the indicated times with SB202190 (2.5 μM) in medium containing 0.5% FBS. Total RNA was isolated and subjected to Northern blotting to determine amounts of LDL receptor, squalene synthase (SS) genes, and actin mRNAs. Autoradiographs were quantitated densitometrically. LDL receptor mRNA levels were normalized to actin mRNA levels. In the bottom panel, results are expressed as the fold induction by SB 202190 as compared with uninduced cells (set at 1). Values obtained are the averages of three separate experiments, with the standard deviations shown.
  • FIG. 2 shows that SB202190 induces LDL receptor expression in a sterol-sensitive manner irrespective of concentrations. HepG2 cells were grown as described in FIG. 1 On [0018] day 4, medium was changed to either 10% LPDS alone or 10% LPDS supplemented with 25-hydroxycholesterol (2 μg/ml) and cholesterol (10 μg/ml), and after 2 hours, cells were treated with indicated concentrations of SB202190 for an additional 4 hours. Levels of LDL receptor and SS mRNAs were determined by Northern analysis and densitometry as described previously. RNA levels were normalized by comparison with levels of actin. Values obtained from cells cultured in the absence of sterols and SB202190 were set at 1. Values shown are the average of two different experiments. The experiment was repeated two times with similar results.
  • FIG. 3 shows activation of p38[0019] MAPK α-isoform interferes with LDL receptor promoter activity. (A) HepG2 cells were co-transfected with LDL receptor-luciferase reporter (plasmid A) (46) and expression vectors encoding MKK6b, MKK6b(A), MKK6b(E), p38MAPK α-isoform, p38MAPK β-isoform, p38MAPK α-isoform (AF) as indicated. Following transfection, cells were washed three times with PBS and maintained in 0.5% FBS. After 16 hours, cells were harvested and luciferase activity was determined and normalized to the protein content of each extract. Luciferase activity expressed by cells transfected with empty vector was given an arbitrary value of 1. The results are presented as means±standard errors and represent at least four individual experiments. The amount of DNA used were: plasmid A (0.75 μg/well), MKK6b, MKK6b(A), MKK6b(E) (0.6 μg/well), p38MAPK (x-isoform, cc(AF) mutant, p38MAPK β-isoform (0.2 μg/well).
  • FIG. 4 shows SB202190-induced LDL receptor expression is mediated by p42/44[0020] MAPK signaling cascade. FIG. 4A shows kinetics of p42/44MAPK, and MEK-1/2 activation by SB202190. 2×105 cells were grown and treated as described in FIG. 1. After the indicated times, equal amounts of cell lysates were blotted with anti-phospho-p42/44MAPK antibody, phosphorylation-independent p42/44MAPK antibody, anti-phospho p46/54JNK antibody, anti-phospho-MEK-1/2, or phosphorylation-independent MKP-1.
  • FIG. 4B shows that HepG2 cells were grown as described in FIG. 1, and were either untreated or treated with SB202190 (2.5 μM) for 4 h either in the absence or presence of indicated concentrations of PD98059 that was added 30 min prior to SB202190 (2.5 μM) addition. Total RNA was subjected to Northern blot analysis, and the filter was hybridized with a [0021] 32P-labeled LDL receptor probe. Values shown are the averages of two different experiments. Ethidium bromide staining of RNA gel before blotting onto a nitrocellulose to demonstrate equal loading of RNA in all lanes. Values obtained from the control cells grown in the absence of SB202190 or PD98059 were arbitrarily set at 1.
  • FIG. 4C shows that cells were grown as described in FIG. 2. On [0022] day 4, medium was changed to either 10% LPDS alone or 10% LPDS supplemented with three different concentrations of 25-hydroxycholesterol (2 μg/ml, 5 μg/ml, or 10 μg/ml) together with cholesterol (10 μg/ml). After 2 h, cells were treated with 2.5 μM SB202190 for an additional 4 h. Cell extracts were prepared, and equal amounts were subjected to SDS-PAGE and immunoblotted with anti-phospho-p42/44MAPK, or phosphorylation-independent anti-p42/44MAPK. Results shown are indicative of three separate experiments.
  • FIG. 5 shows that SB202190 induces LDL receptor expression and phosphorylation of p42/44[0023] MAPK in HeLa cells, and pretreatment with PD98059 inhibited SB20219-induced LDL receptor expression. FIG. 5A shows that 1×105 cells were plated on day 0, and were refed with fresh medium on day 2. On day 4, cells were treated with SB202190 (2.5 μg/ml) in 0.5% FBS. After the indicated times, cells were lysed in SDS sample buffer, and subjected to Western blotting with anti-phospho-p42/44MAPK or phosphorylation-independent anti-p42/44MAPK. Equal amounts of p38MAPK was observed in all the lanes (data not shown).
  • FIG. 5B shows that HeLa cells were either untreated or treated with SB202190 (2.5 μM) for 4 h either in the absence or presence of of PD98059 (5 μM). PD98059 was added 30 min prior to addition of SB202190. After the indicated times, total RNA was isolated and subjected to Northern blotting. RNA gel stained with ethidium bromide before blotting onto nitrocellulse is shown to demonstrate equal loading of RNA. [0024]
  • FIG. 6 shows modulation of p42/44[0025] MAPK activity does not affect p38MAPK phosphorylation in response to a variety of stress-inducers in HepG2-ΔRaf-1:ER cells. Cells were either left untreated or stimulated with 1 μM ICI 182,780 for 1.5 h, and then treated with either anisomycin (50 ng/ml) or IL-1β (5 ng/ml) for 30 min. Cells were then lysed in SDS sample buffer, and equal amounts of whole cell lysates were separated by SDS-PAGE in 10% gels. Activated MAPKs levels were analyzed by blotting using anti-phospho-p42/44MAPK, and anti-phospho-p38MAPK. Protein levels were analyzed by using phosphorylation-independent anti-p42/44MAPK following electrotransfer of total proteins onto nitrocellulose. Similar results were obtained in three separate experiments.
  • FIG. 7 shows that SB202190-induced LDL receptor expression does not require new protein synthesis. HepG2 cells were grown as described in FIG. 1. On [0026] day 4, cells cultured for 1 h in 0.5% FBS containing different amounts of cycloheximide (1 μg/ml, 2 μg/ml, and 5 μg/ml) were then incubated with SB202190 (2.5 μM) for an additional 4 h. LDL receptor and actin mRNA levels were analyzed by Northern blotting of total RNA. Values obtained from cells with no additions were arbitrarily set at 1. The experiment was repeated two times with similar results. Results shown are representative of one experiment.
  • FIG. 8 shows effects of different protein kinase C inhibitors on SB202190-induced LDL receptor expression in HepG2 cells. In FIG. 8A, HepG2 cells were pretreated for 30 minutes with the indicated concentration of the above inhibitors and then treated with 2.5 μM SB202190 for an additional 4 hours. Total RNA was subjected to Northern blotting for measurement of LDL receptor mRNA levels. Ethidium bromide staining of RNA gel before blotting onto a nitrocellulose paper to demonstrate equal loading of RNA in all lanes. Similar results were obtained in four seaparate experiments. [0027]
  • FIG. 8B shows that staurosporine blocked SB202190-induced p42/44[0028] MAPK and MEK-1/2 activation. HepG2 cells were cultured in the absence or presence of 1 μM staurosporine or 5 μM PD98059 for 30 min. SB202190 (2.5 μM) was then added to the cells for 4 h, cell lysate was subjected to immunoblotting to measure the phosphorylation levels of p42/44MAPK and MEK-1/2.
  • FIG. 9 shows a suggested model for cross-talk between p38[0029] MAPK and p42/44MAPK signaling pathways for regulating LDL receptor expression.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention examines the role of p38[0030] MAPK in the regulation of the LDL receptor expression and demonstrates that there is a cross-talk between p42/44MAPK and p38MAPK signaling cascades, and p38MAPK negatively regulates LDL receptor expression via the p42/44MAPK signaling cascade.
  • The present invention reports that SB202190 alone, a specific inhibitor of p38[0031] MAPK, induces low density lipoprotein (LDL) receptor expression (6-8 fold) in a sterol-sensitive manner in HepG2 cells. Consistent with this finding, selective activation of the p38MAPK signaling pathway by expression of MKK6b (E), a constitutive activator of p38MAPK, significantly reduced LDL receptor promoter activity. Expression of the p38MAPK α-isoform had a similar effect, whereas, expression of the p38MAPK βII-isoform had no effect on LDL receptor promoter activity. SB202190-dependent increase in LDL receptor expression was accompanied by induction of p42/44MAPK and inhibition of this pathway completely prevented SB202190-induced LDL receptor expression, suggesting that p38MAPK negatively regulates the p42/44MAPK cascade and the responses mediated by this kinase. Cross-talk between these kinases appears to be one-way because modulation of p42/44MAPK activity did not affect p38MAPK activation by a variety of stress-inducers. This cross-talk is independent of protein synthesis and clearly requires signaling in a protein kinase C-independent manner via a staurosporine-sensitive protein kinase that lies upstream of MEK-1/2. Taken together, these findings reveal a hitherto unrecognized one-way communication that exists between p38MAPK and p42/44MAPK and provide the first evidence that through the p42/44MAPK signaling cascade, the p38MAPK α-isoform negatively regulates LDL receptor expression, representing a novel mechanism of fine-tuning cellular levels of cholesterol in response to a diverse set of environmental cues.
  • In one embodiment of the present invention, there is provided a compound for inducing low density lipoprotein receptor expression, wherein the compound is a p38[0032] MAPK inhibitor or a compound that activates p42/44MAPK. The representative examples of such compounds are SB202190 and SB203580.
  • In another embodiment of the present invention, there is provided a method of inducing LDL receptor expression in a cell by administering to the cell with the compound disclosed herein. The cell is either hepatic or nonhepatic. Preferably, the compound induces the LDL receptor expression by 6-8 fold and further reduces cholesterol level in the cell. [0033]
  • In still another embodiment of the present invention, there is provided a method of treating an individual having hypercholesterolemia by administering the compound disclosed herein. Preferably, the compound is administered at a concentration range of from about 1 μM to about 100 μM. [0034]
  • The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion. [0035]
  • EXAMPLE 1
  • Materials [0036]
  • Cycloheximide, calphostin C, PD98059, and SB202190 were purchased from Calbiochem (San Diego, Calif.). Phospho-specific antibodies to the activated forms of p42/44[0037] MAPK (Thr 202/Tyr 204), p46/54JNK (Thr 183/Tyr 188), p38MAPK (Thr 180/Tyr 182), and MEK-1/2 (Ser 217/221) were purchased from New England Biolabs (Beverly, Mass.). Antibodies to p42/44MAPK, mitogen-activated protein kinase phosphatase-1 (MKP-1) were obtained from Santa Cruz Biotechnology (Santa Cruz, Calif.). IL-1β and TNF were purchased from R & D Systems Inc. (Minneapolis, Minn.). TRIzol and all tissue culture supplies were from Life Technologies Inc. (Gaithersburg, Md.). Zeta probe blotting membrane and the protein assay reagent were purchased from Bio-Rad (Hercules, Calif.). [α-32P]dCTP (3000 Ci/mmol) was obtained from Dupont (Boston, Mass.), and the enhanced chemiluminescence (ECL) detection kit was obtained from Amersham International (Arlington Heights, Ill.). pSV-β-galactosidase (pSV-β-Gal) vector was purchased from Promega, and was used as a positive control for monitoring transfection efficiencies of HepG2 cells (24). Dual-Light chemiluminescent reporter gene assay system for the combined detection of luciferase and β-galactosidase was purchased from TROPIX, Inc.
  • EXAMPLE 2
  • Cell Culture [0038]
  • Human hepatoma cell line HepG2 and its derivative HepG2-ΔRaf-1:ER cell line that stably expresses the ΔRaf-1:ER chimera were maintained as monolayer cultures in a humidified 5% CO[0039] 2 atmosphere at 37° C. in Eagle's minimum essential medium (EMEM) (BioWhittaker, MD) supplemented with 10% fetal bovine serum (FBS) (Life Technologies Inc.), 2 mM L-glutamine, 20 units/ml penicillin and 20 μg/ml streptomycin sulphate.
  • EXAMPLE 3
  • Immunoblot Analysis [0040]
  • Proteins were fractionated by SDS-PAGE with an 10% acrylamide separation gel, and the separated proteins were transferred to nitrocellulose, and processed as described previously (43, 44). Briefly, membranes were incubated in 20 mM Tris-HCl, pH 7.6, 137 mM NaCl, 0.2% v/v Tween-20 (Tris/NaCl/Tween-20) with 5% w/v non-fat dried milk for 1 h, washed in Tris/NaCl/Tween-20 (3×5 min), and incubated for 1 h with primary antibody in Tris/NaCl/Tween-20 containing 1% milk at room temperature for non phospho-specific antibodies and overnight at 4° C. for phospho-specific antibodies. The following dilutions were used for individual antibodies against different proteins: p42/44[0041] MAPK (1:1600); phospho-p42/44MAPK (1:1000); phospho-p46/54JNK (1:1000); phospho-p38MAPK (1:1600); phospho-MEK-1/2 (1:1000); MKP-1 (1:1500). After further washing in Tris/NaCl/Tween-20, membranes were incubated for 1 h with horseradish peroxidase-linked anti-IgG secondary antibody (Bio-Rad, diluted 1:5000), and immunoreactive proteins were detected by ECL as described by the supplier. Quantitative analyses of protein levels were performed by densitometric scanning of the autoradiograms, and are representative of 3 or more independent experiments.
  • EXAMPLE 4
  • Northern Analysis [0042]
  • Total RNA was isolated using TRIzol, and Northern blotting was done essentially as described earlier ([0043] 43, 44). Briefly, 20 μg total cellular RNA was fractionated on 1% formaldehyde agarose gel and transferred to Zeta Probe membrane by capillary blotting. RNA blots were hybridized with LDL receptor and squalene synthase specific single-stranded M13 probes labeled with [α-32P]dCTP. Hybridized filters were washed and exposed to Kodak X-ray film. The relative intensities of specific bands were determined densitometrically within the linear range of the film on a model 300A laser densitometer (Molecular Dyanamics, CA) with Image Quant software. LDL receptor mRNA was normalized to squalene synthase (SS) (included in this study) or to β-actin (data not shown) mRNA level, and data for each point were plotted as the percentage of LDL receptor mRNA as compared to controls.
  • EXAMPLE 5
  • Expression Vectors and Reporter Constructs [0044]
  • Expression vectors of MKK6b, MKK6b(E) (a double mutant of MKK6b in which serine[0045] 207 and threonine211 are replaced with glutamic acid), MKK6b(A) (a mutant of MKK6b in which serine222 is replaced with alanine), p38MAPK α, p38MAPK α(AF) (a double mutant of p38MAPK α-isoform in which threonine188 and tyrosine190 with phenylalanine), and p38MAPK βII have been described elsewhere (19, 26). Reporter gene of LDL receptor promoter-luciferase (plasmid A), in which the 5′-flanking region of human LDL receptor promoter was fused to firefly luciferase gene has also been described previously (45, 46).
  • EXAMPLE 6
  • Transient Transfection and Luciferase Assay [0046]
  • HepG2 cells were transfected by the Lipofectamine method. For the LDL receptor promoter-luciferase expression assays, HepG2 cells were seeded at a density of 1×10[0047] 6 cells per 6 well plates and co-transfected with 0.25 μg of relevant expression vector, or the corresponding empty vector (45, 46). Luciferase activity was measured 12-18 h after transfection. Normalization was achieved by co-transfecting 0.1 μg of pSV-β-Gal, a β-galactosidase reporter construct as an internal control for the transfection efficiency. Luciferase and P-galactosidase activities were measured according to the TROPIX protocol. Data are representive for at least three independent experiments performed in duplicate and are expressed as “fold increase in luciferase activity,” which was calculated relative to the basal level of LDL receptor promoter reporter activity (set to 1 unit) and corrected for empty vector effects for each expression vector.
  • EXAMPLE 7
  • The Specific p38[0048] MAPK Inhibitor SB202190 by Itself Induces LDL Receptor Expression
  • To examine whether the p38[0049] MAPK is involved in regulation of LDL receptor expression, the effects of inhibition of this kinase was first examined by using a specific inhibitor. The activity of p38MAPK was inhibited by using SB202190, a highly selective inhibitor of p38MAPK that does not affect the activity of other relevant kinases, even at high concentrations. HepG2 cells were treated with SB202190 for various times and the effect on LDL receptor expression was measured by Northern blotting. SB202190 induces LDL receptor expression in a time-dependent manner without significantly affecting expression of a house-keeping actin gene or another sterol-sensitive gene of the cholesterol biosynthetic pathway, SS (FIG. 1). A significant increase in LDL receptor expression was apparent at 4 h and a plateau was reached after 8 h treatment of SB202190. Another specific p38MAPK inhibitor SB203580 also induced LDL receptor expression, whereas the inactive derivative SB202474 did not induce LDL receptor expression (data not shown). The most effective concentrations were 2.5 μM for SB202190 and 10 μM for SB203580.
  • Because LDL receptor is negatively regulated by sterols, whether SB202190 can induce LDL receptor expression in the presence of sterols was then tested. As shown in FIG. 2, presence of sterols suppressed SB202190-induced LDL receptor expression. However, a slight increase in the suppressed levels of LDL receptor was observed in a dose-dependent manner on treatment with SB202190. [0050]
  • Because pyridinyl imidazoles that are closely related to SB203580 have cyclooxygenase-inhibitory activity (47, 48), experiments were conducted to determine whether blockade of this activity with indomethacin (49) can alter LDL receptor expression. Importantly, unlike SB202190, 10 μM indomethacin did not significantly alter LDL receptor expression (results not shown). From the above data, it seemed probable that the effect of SB202190 on LDL receptor expression is related to its inhibition of p38[0051] MAPK.
  • EXAMPLE 8
  • The p38[0052] MAPK Pathway Negatively Regulates LDL Receptor Expression
  • To investigate the role of p38[0053] MAPK signaling pathway in the regulation of LDL receptor expression more directly, previously characterized expression constructs was used to modulate either positively or negatively the endogenous p38MAPK activity. LDL receptor transcription was monitored by transfecting HepG2 cells with a previously cloned fragment of the human LDL receptor promoter fused to the luciferase reporter gene (46), together with the relevant constructs. In several cell systems, p38MAPK activity can be stimulated by coexpression of p38MAPK with MKK6b, or by expression of constitutively activated MKK6b(E), in which the activating phosphorylation residues Ser207 and Thr211 were replaced by glutamic acids (19, 50). Expression of MKK6b(E) reduced luciferase gene expression many fold, when compared with the luciferase expression in control cells transfected with the empty vector (FIG. 3). This dramatic down-regulation of the LDL receptor reporter expression in cells overexpressing the constitutively active MKK6b mutant most likely resulted from a decrease in p42/44MAPK activity in these cells, as assessed by measuring activity of the co-transfected p42/44MAPK plasmid (FIG. 3). Conversely, expression of the dominant-negative regulatory MKK6b(A) construct slightly increased LDL receptor-luciferase expression, correlating with a partial inhibition of endogenous MKK6b activity in cells expressing this mutated form of MKK6b.
  • To rule out the possibility that MKK6b(E) may suppress LDL receptor promoter activity in a p38[0054] MAPK-independent manner, it was tested whether MKK6b(E)-inhibited LDL receptor expression was due to activation of p38MAPK. HepG2 cells were transfected with the human LDL receptor promoter-luciferase reporter gene along with expression vectors encoding p38MAPK α or β(βII)-isoform, MKK6b, or empty vector. It is interesting to note that expression of p38MAPK α-isoform or with MKK6b alone was able to suppress reporter gene expression mildly. However, coexpression of p38MAPK α-isoform and MKK6b together strongly reduced luciferase activity manyfold (FIG. 3). The effect of MKK6b is dependent on p38MAPK activation, since MKK6b failed to stimulate reporter gene activity when it was co-transfected with an inactive p38MAPK α(AF) mutant, in which one of the activating phosphorylation residues, Tyr 182, was replaced by phenylalanine. At the same time, transfection of p38MAPK β-isoform alone or with MKK6b had no significant effect. These results demonstrate that activation of p38MAPK by itself is sufficient to suppress reporter gene expression. Furthermore, it is concluded that p38MAPK α-isoform and not p38MAPK β-isoform mediates SB202190-induced LDL receptor expression.
  • EXAMPLE 9
  • SB202190-Induced LDL Receptor Expression is Mediated by p42/44[0055] MAPK Signaling Cascade
  • Since p42/44[0056] MAPK have been shown to play a critical role in induction of LDL receptor expression (43-45), it is determined whether inhibition of p38MAPK by SB202190 leads to activation of other mitogen-activated protein kinases, p42/44MAPK and p46/54JNK. HepG2 cells were incubated with SB202190, and the activity of p42/44MAPK and p46/54JNK was determined by immunoblot analysis with antibodies that recognized the activated phosphorylated forms of these kinases in cell extracts obtained at different times.
  • In contrast to the rapid and transient growth factor-induced activation of p42/44[0057] MAPK, SB202190 treatment caused a delayed and prolonged activation with no effect on p42/44MAPK protein levels (FIG. 4A). Weak activation was observed at 1 hour after treatment, and maximal activation was obtained at 4-8 hours after stimulation. The activated p42/44MAPK remained elevated at least until 24 hours, the maximum time point measured. Furthermore, changes in p42/44MAPK activity took place with little or no effect on the activity or levels of p46/54JNK (FIG. 4A). Because the kinetics of MEK-1/2 phosphorylation paralleled the appearance of p42/44MAPK, it is likely that MEK-1/2 is upstream of p42/44MAPK. Because MEK-1/2 directly phosphorylates and activates p42/44MAPK, PD98059 was used to determine the role of this pathway in SB202190-induced LDL receptor expression. PD98059 inhibited both SB202190-induced p42/44MAPK phosphorylation (FIG. 4B) and induction of LDL receptor expression, suggesting that LDL receptor induction is mediated by SB202190-dependent p42/44MAPK activation. Furthermore, lack of effect of sterols on SB202190-induced p42/44MAPK phosphorylation ruled out involvement of a sterol-sensitive step in p42/44MAPK activation (FIG. 4C). These results suggested that p38MAPK negatively regulates p42/44MAPK activity and the SB202190-dependent increase in LDL receptor expression is mediated by p42/44MAPK.
  • It was then tested whether p38[0058] MAPK inhibition by SB202190 induces LDL receptor expression in other cell types. In HeLa cells, SB202190 treatments induced p42/44MAPK activity and caused a increase in LDL receptor expression (FIG. 5). One interesting feature is the delayed and sustained kinetics of p42/44MAPK activation by SB202190 in HepG2 cells as compared to rapid and transient kinetics of p42/44MAPK increase in non-hepatic HeLa cells (FIG. 5). It is possible that the differences in the kinetics and extents of stimulation of p42/44MAPK activity and LDL receptor induction may reflect the underlying differences between these cell types. Inspite of these differences, consistent with the role of p42/44MAPK, PD98059 blocked SB202190-induced LDL receptor expression.
  • EXAMPLE 10
  • Modulation of Endogenous p42/44[0059] MAPK Activity Does Not Affect p38MAPK Activation
  • Whether inhibition or activation of p42/44[0060] MAPK regulates p38MAPK activation by stress-inducers was determined next. To test this possibility, a HepG2-derived cell line (HepG2ΔRaf:ER) expressing an estradiol-dependent human Raf-1 protein kinase was generated. In this cell line, the ΔRaf-1:ER chimera is activated in response to estradiol or anti-estrogen ICI 182780, thereby activating MEK-1/2 and then p42/44MAPK (51, 52).
  • Addition of ICI 182780 to these cells stimulated p42/44[0061] MAPK within minutes (FIG. 6). The p42/44MAPK activity increased for up to 1 h, and remained elevated in the presence of ICI 182780. This cell system was used to directly measure the effect of ICI 182780 on p38MAPK activation by a variety of stress-inducers. As shown in FIG. 6, no significant effects was observed on p38MAPK activation by anisomycin or IL-1ε on super-induction of p42/44MAPK activity with ICI 182780. Likewise, inhibition of p42/44MAPK with PD98059 has no effect on activity of other MAPKs (data not shown). This is consistent with earlier demonstration that PD98059 failed to block p38MAPK activation in response to IL-1β or TNF at concentrations that completely blocked p42/44MAPK activation by MEK-1/2 (43). Similar results have also been obtained by Frasch et al. in human neutrophils (53). Collectively, the above results demonstrate that p42/44MAPK does not regulate p38MAPK activation by a variety of transcriptional modulators.
  • EXAMPLE 11
  • SB202190-Induced LDL Receptor Expression is Independent of New Protein Synthesis [0062]
  • To determine whether the induction of LDL receptor expression by SB202190 may require new protein synthesis, HepG2 cells were incubated with SB202190 in the absence or presence of different concentrations of cycloheximide. Data shown in FIG. 7 demonstrates that inhibition of translation has no effect on LDL receptor expression, suggesting that an additional protein synthesis is not required to stimulate LDL receptor expression. [0063]
  • EXAMPLE 12
  • Involvement of a Staurosporine-Sensitive Step in SB202190-Induced p42/44[0064] MAPK Activation and Induction of LDL Receptor Expression
  • There may be several pathways leading to p42/44[0065] MAPK activation. Protein kinase C (PKC) is implicated in at least one of them because phorbol-ester, a well-known activator of protein kinase C, activates p42/44MAPK and inhibitors of protein kinase C prevent agonist-induced p42/44MAPK activation in several types of cells (44).
  • To assess the involvement of protein kinase C, HepG2 cells pretreated with various protein kinase C inhibitors were subjected to SB202190 treatments. It is interesting to note that highly selective protein kinase C inhibitors, calphostin C, Bis I, and Go6976 has no effect on SB202190-induced LDL receptor expression (FIG. 8A). Conversely, staurosporine, a non-specific inhibitor of various protein kinases, including protein kinase C, completely prevented induction of LDL receptor expression by SB202190. As expected, pretreatment of HepG2 cells with staurosporine blocked SB202190-induced P42/44[0066] MAPK and MEK-1/2 activation, further supporting the involvement of a staurosporine-sensitive step in p42/44MAPK activation (FIG. 8B).
  • Discussion [0067]
  • Although p38[0068] MAPK and p42/44MAPK are members of different mitogen-activated protein kinase subfamilies, the present inevntion provides an evidence for one-way cross-talk between these mitogen-activated protein kinases. The conclusions that SB202190-induced LDL receptor expression is due to inhibition of p38MAPK, and this pathway exerts its effect on LDL receptor expression through p42/44MAPK activation, is based on the following observations. First, under experimental conditions where PD98059 completely inhibited phosphorylation of p42/44MAPK, this MEK-1/2 inhibitor blocked SB202190-induced LDL receptor expression. The selectivity of PD98059 has been described in several studies, and the possibility that SB202190 inhibits another p38MAPK is unlikely because this inhibitor is without effect even at higher concentrations (100 μM) on either p46/54JNK, or p42/44MAPK, or multiple other related protein kinases including the homologues p38MAPK γ (SAPK3) and SAPK4, which share 60% identity with p38MAPK αand β.
  • The present invention also demonstrated that both PD980569 and SB202190 failed to block other mitogen-activated protein kinases in vivo at concentrations that completely blocked p38[0069] MAPK activity (43). Furthermore, to rule out the possibility that SB202190 may inhibit other unknown targets to fully execute its effect on p42/44MAPK and LDL receptor expression, it is shown that expression of MKK6b(E), the constitutive activator of p38MAPK, significantly suppressed LDL receptor promoter activity (FIG. 3). The effect of MKK6b(E) was dependent on activation of p38MAPK, since coexpression of MKK6b with p38MAPK, but not the inactive p38MAPK (AF) mutant, inhibited LDL receptor expression. Finally, consistent with the role of p42/44MAPK in SB202190-induced LDL receptor expression, inhibition of p38MAPK resulted in induction of its activity in a time-dependent manner. Activation of both the p42/44MAPK and LDL receptor expression was observed with similar kinetics suggesting that the activated p42/44MAPK is required to continuously increase the expression of the LDL receptor gene. p42/44MAPK phosphorylation started at 4 hours, with a maximal activation observed after 8 hours, and remained persistent up to 24 hours following treatment with SB202190. This delay in SB202190-induced p42/44MAPK is most likely due to the late appearance of an immediate or downstream substrate of the p38MAPK pathway either through de novo synthesis, post-translational modification or localization. The finding that SB202190-induced LDL receptor induction is not inhibited by cycloheximide (FIG. 7), a protein synthesis inhibitor, rules out de novo synthesis. It is more likely that induction of p42/44MAPK by SB202190 requires post-translational modification of the already existing substrate(s). Moreover, the lack of effect of sterols on SB202190-induced p42/44MAPK activation supports the notion that the sterol-sensitive step either lie downstream of p42/44MAPK or is a part of an independent signaling pathway.
  • This study represents the first demonstration of a functional difference observed for p38[0070] MAPK isoforms in the regulation of gene expression. Recent evidence indicates the existence of at least four distinct isoforms of p38MAPK; p38MAPK α, also named CSBP2 or stress-activated protein kinase 2a; p38MAPK β, also named stress-activated protein kinase 2b and its splice isoform β2, p38MAPK γ, also termed stress-activated protein kinase 3 or ERK6; and p38MAPK δ, otherwise known as stress-activated protein kinase 4. The four isoforms are similar in size, show about 60 to 75% sequence homology, and are all activated by TNF, IL-1, ultraviolet radiation, and hyperosmolar medium. Some isoforms show a pronounced preference in tissue expression and selective interaction with upstream kinases and downstream substrates, pointing to highly specialized functions. These isoforms also differ in susceptibility to inhibition by SB202190; both α- and β-isoforms of p38MAPK are inhibited by SB202190, whereas the γ and β isoforms are insensitive (26). Therefore, it seems less likely that either the γ or the δ isoform is responsible for the induction of LDL receptor expression by SB202190.
  • Since hepatic cells contain similar levels of p38[0071] MAPK α- and β-isoforms, next question to be asked is which of the isoform is responsible for the SB202190-induced LDL receptor expression in HepG2 cells. Availability of expression vectors encoding the individual p38MAPK isoforms made it possible to directly test their involvement by co-transfection studies. It was found that, unlike p38MAPK β-isoform, expression of the p38MAPK α-isoform suppressed LDL receptor promoter activity. These studies suggest that SB202190-induced LDL receptor expression is mediated by the inhibition of α-isoform of p38MAPK. The observed difference in the role of α- and β-isoforms is consistent with a recent report that showed opposite effects of these isoforms on SB202190-induced apoptosis (54).
  • At this point, it is not clear why the a- and not the β-isoform negatively controls LDL receptor expression via p42/44[0072] MAPK. One possibility is that these two p38MAPK isoforms may differ in their substrate specificity, and such differences could allow coupling of p38MAPK α-isoform to the p42/44MAPK signaling pathway. In an attempt to elucidate the mechanism underlying this cross-talk, various inhibitors of protein kinase C were used. It is interesting to note that staurosporine completely blocked SB202190-induced p42/44MAPK activation (FIG. 8). Staurosporine was originally considered to be a specific inhibitor of protein kinase C, but further studies revealed it also inhibits a variety of tyrosine kinases as well as serine-threonine kinases (55).
  • The role of protein kinase C in SB202190-induced LDL receptor expression is ruled out by the lack of effects of specific PKC inhibitors on the induction process. At this point, it is not clear which protein kinase is inhibited by staurosporine, but it is safe to predict that cross-talk between p42/44[0073] MAPK and p38MAPK pathways clearly requires signaling via a staurosporine-sensitive protein kinase that lie upstream of MEK-1/2. Because the number of substrates for p38MAPK that have been characterized in any system is few, and those that might contribute to activation of p42/44MAPK pathways have not been explored, this remains a question for future endeavors.
  • The suppression of p42/44[0074] MAPK activity that is initiated by p38MAPK activation provides a critical link in the signaling events preceding apoptosis. The ability of stressful signals to stimulate p38MAPK activity has led to the suggestion that this pathway may function to communicate growth inhibitory and apoptotic signals within the cell. p38MAPK is involved in the regulation of apoptosis, since overexpression of kinases that can activate p38MAPK resulted in the induction of apoptosis (56, 57), and inhibition of p38MAPK activity has been shown to suppress apoptosis (58). However, simple transient activation of the stress kinase cascades is not always sufficient to induce apoptosis. For example, TNF promotes a significant induction of p46/54JNK and p38MAPK but does not invariably induce apoptosis through induction of caspases (59). In this regard, it was suggested that concomitant inactivation of survival signals may be a prerequisite for p46/54JNK and p38MAPK to induce cell death (60). Interestingly, deprivation of neurotrophic factors in PC-12 cells or ultraviolet-irradiation of NIH-3T3 cells not only activates the stress kinase cascades but also leads to a dramatic inhibition of the p42/44MAPK pathway (56, 61). In fact, overexpression of p42/44MAPK in NIH-3T3 cells impaired a large part of the ultraviolet-induced apoptotic response (61). Furthermore, inhibition of p42/44MAPK alone has been shown to induce apoptosis through activation of caspases (62).
  • The negative regulation of p42/44[0075] MAPK by the p38MAPK is consistent with the above results and provides a critical link between the p38MAPK activation and the concomitant inhibition of p42/44MAPK signaling cascade. It is likely that induction of apoptosis due to p38MAPK activation could be at least partly due to inhibition of the p42/44MAPK. The contradiction regarding the role of p38MAPK in apoptosis may be due to differences in the degree and extent of cross-talk between the “death” signal mediated by p38MAPK and the survival signal generated by activation of p42/44MAPK, and the differential responses may lead to outcome in a cell- and stimuli-specific manner. Such a mechanism may reconcile the contradictory roles that have been suggested for p38MAPK in apoptosis.
  • In conclusion, while studying the role of p38[0076] MAPK in the regulation of LDL receptor expression, a one-way inhibitory cross-talk was unraveled between the p42/44MAPK and p38MAPK signaling pathways. In conjunction with earlier work (43-45), the above results demonstrate that both p38MAPK and p42/44MAPK signaling pathways are important in regulating LDL receptor expression in intact cells. A signal transduction cascade involving p42/44MAPK and p38MAPK has been proposed to account for SB202190-induced LDL receptor induction by a staurosporine-sensitive protein kinase (FIG. 9). The signaling cascade leading to p42/44MAPK activation is still not known and might involve a complex combinatorial, spatial cross-talk of already synthesized messenger molecules. The interplay between these signaling cascades should be an important process and dynamic balance may be critical for determining the outcome of a wide array of biological processes. Elucidation of the signaling components involved in this communication will significantly advance the ability to design novel strategies for the treatment of hypercholesterolemia and for understanding other important pathophysiological processes.
  • The following references were cited herewith. [0077]
  • 1. Marshall, C. J. (1995) [0078] Cell 80: 179-185
  • 2. Seger, etval., [0079] FASEB (1995) 9,726-735
  • 3. Whitmarsh, et al., (1996) [0080] J. Mol. Med. 74: 589-607
  • 4. Robinson, et al., (1997) [0081] Curr. Opin. Cell Biol. 9, 180-
  • 5. Davis, (1994) [0082] Trends Biochem. Sci. 19: 470-473
  • 6. Cano, et al., (1995) [0083] Trends Biochem. Sci. 20, 117-122
  • 7. Cobb, et al., (1995) [0084] J. Biol. Chem. 270: 14843-14846
  • 8. Khokhlatchev, et al., (1997) [0085] J. Biol. Chem. 272: 11057-11062
  • 9. Wood, et al., (1992) [0086] Cell 68: 1041-1050
  • 10. Howe, et al., (1992) [0087] Cell 71: 335-342
  • 11. Kyriakis, et al., (1994) [0088] Nature 369: 156-160
  • 12. Derijard, et al., [0089] Cell 76: 1025-1037
  • 13. Minden, et al., (1994) [0090] Mol. Cell. Biol. 14: 6683-6688
  • 14. Raingeaud, et al., (1995) [0091] J. Biol. Chem. 270: 7420-7426
  • 15. Han, et al., (1994) [0092] Science 265: 808-811
  • 16. Gupta, et al., (1996) [0093] EMBO J. 15: 2760-2770
  • 17. Lee, et al., (1994) [0094] Nature 372, 739-746.
  • 18. Rouse, et al., (1994) [0095] Cell 78: 1027-1037.
  • 19. Jiang, et al., (1996) [0096] J. Biol. Chem. 271: 17920-17926
  • 20. Lechner, et al., (1996) [0097] Proc. Natl. Acad. Sci. U.S.A. 93: 4355-4359
  • 21. Li, et al., (1996) [0098] Biochem. Biophys. Res. Commun. 228: 334-340
  • 22. Mertens, et al., (1996) [0099] FEBS Lett. 383: 273-276
  • 23. Cuenda, et al., (1997) [0100] EMBO J. 16: 295-305
  • 24. Wang, et al., (1997) [0101] J. Biol. Chem. 272: 23668-23674
  • 25. Goedert, et al., (1997) [0102] EMBO J. 16: 3663-3671
  • 26. Enslen, et al., (1998) [0103] J. Biol. Chem. 273: 1741-1748
  • 27. Derijard, et al., (1995) [0104] Science 267: 682-685
  • 28. Lin, et al., (1995) [0105] Science 268: 286-290
  • 29. Sanchez, et al., (1994) [0106] Nature 372: 794-798
  • 30. Moriguchi, et al., (1996) [0107] J. Biol. Chem. 271: 13675-13679
  • 31. Holland, et al., (1997) [0108] J. Biol. Chem. 272: 24994-24998
  • 32. Raingeaud, et al., (1996) [0109] Mol. Cell. Biol. 16: 1247-1255
  • 33. Treisman, R. (1996) [0110] Curr. Opin. Cell. Biol. 8: 205-215
  • 34. Pang, et al., (1995) [0111] J. Biol. Chem. 270, 13585-13588
  • 35. Cuenda, et al., (1995) [0112] FEBS Lett. 364, 229-233
  • 36. Beyaert, et al., (1996) [0113] EMBO J. 15: 1914-
  • 37. Hazzalin, et al., (1996) [0114] Curr. Biol. 6: 1028-
  • 38. Crawley, et al., (1997) [0115] J. Biol. Chem. 272: 15023-
  • 39. Kawasaki, et al., (1997) [0116] J. Biol. Chem. 272: 18518-
  • 40. Graves, et al., (1998) [0117] J. Immunol. 161: 168-
  • 41. Tan, et al., (1996) [0118] EMBO J. 15: 4629-
  • 42. Iordanov, et al., (1997) [0119] EMBO J. 16: 1009-
  • 43. Kumar, et al., (1998) [0120] J. Biol. Chem. 273: 15742-15748
  • 44. Kumar, et al., (1997) [0121] J. Lipid Res. 38, 4220-4228
  • 45. Mehta, et al., (1999) [0122] J. Lipid Res., Manuscript submitted
  • 46. Mehta, et al., (1996) [0123] J. Biol. Chem. 271: 33616-33622
  • 47. Kramer, et al., (1996) [0124] J. Biol. Chem. 271: 27723-27729
  • 48. Lee, et al., (1993) [0125] Ann. N.Y. Acad. Sci. 696: 149-170
  • 49. Vane, J. R. (1971) [0126] Nat. New. Biol. 231: 232-235
  • 50. Han, et al., (1996) [0127] J. Biol. Chem. 271: 2886-2891
  • 51. Bosch, et al., (1997) [0128] Oncogene 15: 1021-1033
  • 52. Woods, et al., (1997) [0129] Mol. Cell. Biol. 17: 5598-5611
  • 53. Frasch, et al., (1998) [0130] J. Biol. Chem. 273: 8389-8397
  • 54. Nemoto, et al., (1998) [0131] J. Biol. Chem. 273: 16415-15420
  • 55. Meggio, et al., (1995) [0132] Europ. J. Biochem. 234: 317-322
  • 56. Xia, et al., (1995) [0133] Science 270: 126-131
  • 57. Ichijo, et al., (1996) [0134] Science 275: 90-
  • 58. Schwenger, et al., (1997) [0135] Proc. Natl. Acad. Sci. U.S.A. 94: 28692873
  • 59. Liu, et al., (1996) [0136] Cell 87: 565-576.
  • 60. Canman, et al., (1996) [0137] Nature 384: 213-214
  • 61. Berra, et al., (1997) [0138] Mol. Cell. Biol. 17: 4346-4354
  • 62. Berra, et al., (1998) [0139] J. Biol. Chem. 273: 10792-10797
  • Any patents or publications mentioned in this specification are indicative of the levels of those skilled in the art to which the invention pertains. These patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference. [0140]
  • One skilled in the art will readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The present examples along with the methods, procedures, treatments, molecules, and specific compounds described herein are presently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art which are encompassed within the spirit of the invention as defined by the scope of the claims. [0141]

Claims (8)

What is claimed is:
1. A compound for inducing low density lipoprotein receptor expression, wherein said compound is selected from the group consisting of a p38MAPK inhibitor and a compound that activates p42/44MAPK.
2. The compound of claim 1, wherein said compound is selected from the group consisting of SB202190 and SB203580.
3. A method of inducing low density lipoprotein receptor expression in a cell, comprising the step of:
administering the compound of claim 1 to said cell.
4. The method of claim 3, wherein said cell is selected from the group consisting of a hepatic cell and a nonhepatic cell.
5. The method of claim 3, wherein said compound induces said low density lipoprotein receptor expression from about 6-fold to about 8-fold.
6. The method of claim 3, wherein said method further reduces cholesterol level in said cell.
7. A method of treating an individual having hypercholesterolemia, comprising the step of:
administering the compound of claim 1 to said individual.
8. The method of claim 7, wherein said compound is administered at a concentration range of from about 1 μM to about 100 μM.
US10/408,968 1999-04-01 2003-04-08 P38MAPK inhibitor and uses thereof Abandoned US20030207849A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/408,968 US20030207849A1 (en) 1999-04-01 2003-04-08 P38MAPK inhibitor and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12734399P 1999-04-01 1999-04-01
US09/539,584 US6602896B1 (en) 1999-04-01 2000-03-31 p38MAPK inhibitor and uses thereof
US10/408,968 US20030207849A1 (en) 1999-04-01 2003-04-08 P38MAPK inhibitor and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/539,584 Division US6602896B1 (en) 1999-04-01 2000-03-31 p38MAPK inhibitor and uses thereof

Publications (1)

Publication Number Publication Date
US20030207849A1 true US20030207849A1 (en) 2003-11-06

Family

ID=22429624

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/539,584 Expired - Fee Related US6602896B1 (en) 1999-04-01 2000-03-31 p38MAPK inhibitor and uses thereof
US10/408,968 Abandoned US20030207849A1 (en) 1999-04-01 2003-04-08 P38MAPK inhibitor and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/539,584 Expired - Fee Related US6602896B1 (en) 1999-04-01 2000-03-31 p38MAPK inhibitor and uses thereof

Country Status (3)

Country Link
US (2) US6602896B1 (en)
AU (1) AU4064100A (en)
WO (1) WO2000059900A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050271661A1 (en) * 2004-05-17 2005-12-08 Combinatorx, Incorporated Methods and reagents for the treatment of immunoinflammatory disorders

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10669528B2 (en) 2015-06-25 2020-06-02 Children's Medical Center Corporation Methods and compositions relating to hematopoietic stem cell expansion, enrichment, and maintenance
EP3429603B1 (en) 2016-03-15 2021-12-29 Children's Medical Center Corporation Methods and compositions relating to hematopoietic stem cell expansion

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656644A (en) * 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
US5837224A (en) * 1996-01-19 1998-11-17 The Regents Of The University Of Michigan Method of inhibiting photoaging of skin

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656644A (en) * 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
US5837224A (en) * 1996-01-19 1998-11-17 The Regents Of The University Of Michigan Method of inhibiting photoaging of skin

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050271661A1 (en) * 2004-05-17 2005-12-08 Combinatorx, Incorporated Methods and reagents for the treatment of immunoinflammatory disorders

Also Published As

Publication number Publication date
WO2000059900A1 (en) 2000-10-12
AU4064100A (en) 2000-10-23
US6602896B1 (en) 2003-08-05

Similar Documents

Publication Publication Date Title
Singh et al. One-way cross-talk between p38MAPK and p42/44MAPK: inhibition of p38MAPK induces low density lipoprotein receptor expression through activation of the p42/44MAPK cascade
Schnabl et al. TAK1/JNK and p38 have opposite effects on rat hepatic stellate cells
Xagorari et al. Inhibition of LPS‐stimulated pathways in macrophages by the flavonoid luteolin
Coffey et al. c-Jun N-terminal protein kinase (JNK) 2/3 is specifically activated by stress, mediating c-Jun activation, in the presence of constitutive JNK1 activity in cerebellar neurons
Cuenda et al. Activation of stress‐activated protein kinase‐3 (SAPK3) by cytokines and cellular stresses is mediated via SAPKK3 (MKK6); comparison of the specificities of SAPK3 and SAPK2 (RK/p38)
Timokhina et al. Kit signaling through PI 3‐kinase and Src kinase pathways: an essential role for Rac1 and JNK activation in mast cell proliferation
Mazzoni et al. Ras regulates sympathetic neuron survival by suppressing the p53-mediated cell death pathway
Lee et al. Constitutive mTOR activation in TSC mutants sensitizes cells to energy starvation and genomic damage via p53
Utsugi et al. C-Jun-NH2-terminal kinase mediates expression of connective tissue growth factor induced by transforming growth factor-β1 in human lung fibroblasts
Reusch et al. Regulation of Raf by Akt controls growth and differentiation in vascular smooth muscle cells
Sridhar et al. Protein kinases as therapeutic targets
Adler et al. Regulation of JNK signaling by GSTp
Birkenkamp et al. The p38 MAP kinase inhibitor SB203580 enhances nuclear factor‐kappa B transcriptional activity by a non‐specific effect upon the ERK pathway
Cavanaugh et al. Differential regulation of mitogen-activated protein kinases ERK1/2 and ERK5 by neurotrophins, neuronal activity, and cAMP in neurons
Jaffe et al. Leptin promotes motility and invasiveness in human colon cancer cells by activating multiple signal‐transduction pathways
Goedert et al. Activation of the novel stress-activated protein kinase SAPK4 by cytokines and cellular stresses is mediated by SKK3 (MKK6); comparison of its substrate specificity with that of other SAP kinases
Tuyt et al. Extracellular-regulated kinase 1/2, Jun N-terminal kinase, and c-Jun are involved in NF-κB-dependent IL-6 expression in human monocytes
Bagowski et al. Cell‐type specific phosphorylation of threonines T654 and T669 by PKD defines the signal capacity of the EGF receptor
Kim et al. Mixed lineage kinase 3 (MLK3)-activated p38 MAP kinase mediates transforming growth factor-β-induced apoptosis in hepatoma cells
Lahti et al. c-Jun NH2-terminal kinase inhibitor anthra (1, 9-cd) pyrazol-6 (2H)-one reduces inducible nitric-oxide synthase expression by destabilizing mRNA in activated macrophages
Nekrasova et al. PAK4 is required for regulation of the cell‐cycle regulatory protein p21, and for control of cell‐cycle progression
Schultz et al. PMA‐induced activation of the p42/44ERK‐and p38RK‐MAP kinase cascades in HL‐60 cells is PKC dependent but not essential for differentiation to the macrophage‐like phenotype
Ishii et al. Activation of the IκBα kinase (IKK) complex by double-stranded RNA-binding defective and catalytic inactive mutants of the interferon-inducible protein kinase PKR
Bellmann et al. p38-dependent enhancement of cytokine-induced nitric-oxide synthase gene expression by heat shock protein 70
Flores et al. Diacylglycerol kinase inhibition prevents IL-2-induced G1 to S transition through a phosphatidylinositol-3 kinase-independent mechanism

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF ARKANSAS MED SCIS LTL ROCK;REEL/FRAME:022239/0186

Effective date: 20050429