Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 42(4): 1139-50, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23403951

ABSTRACT

Acquired chemoresistance and epithelial-to-mesenchymal transition (EMT) are hallmarks of cancer progression and of increasing clinical relevance. We investigated the role of miRNA and p38 mitogen-activated protein kinase (MAPK) signaling in the progression of breast cancer to a drug-resistant and mesenchymal phenotype. We demonstrate that acquired death receptor resistance results in increased hormone-independent tumorigenesis compared to hormone-sensitive parental cells. Utilizing global miRNA gene expression profiling, we identified miRNA alterations associated with the development of death receptor resistance and EMT progression. We further investigated the role of p38 MAPK in this process, showing dose-dependent inactivation of p38 by its inhibitor RWJ67657 and decreased downstream ATF and NF­κB signaling. Pharmacological inhibition of p38 also decreased chemoresistant cancer tumor growth in xenograft animal models. Interestingly, inhibition of p38 partially reversed the EMT changes found in this cell system, as illustrated by decreased gene expression of the EMT markers Twist, Snail, Slug and ZEB and protein and mRNA levels of Twist, a known EMT promoter, concomitant with decreased N­cadherin protein. RWJ67657 treatment also altered the expression of several miRNAs known to promote therapeutic resistance, including miR­200, miR­303, miR­302, miR­199 and miR­328. Taken together, our results demonstrate the roles of multiple microRNAs and p38 signaling in the progression of cancer and demonstrate the therapeutic potential of targeting the p38 MAPK pathway for reversing EMT in an advanced tumor phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Imidazoles/pharmacology , MicroRNAs/metabolism , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Survival/drug effects , Cell Transformation, Neoplastic , Cluster Analysis , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mice , MicroRNAs/genetics , Transcriptome , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Am J Cancer Res ; 2(4): 446-58, 2012.
Article in English | MEDLINE | ID: mdl-22860234

ABSTRACT

Endocrine therapy resistance is a primary cause of clinical breast cancer treatment failure. The p38 mitogen activated protein kinase (MAPK) signaling pathway is known to promote ligand independent tumor growth and resistance to endocrine therapy. In this study, we investigated the therapeutic potential of the p38 inhibitor RWJ67657 in the treatment of tamoxifen resistant MDA-MB-361 cells. RWJ67657 dose-dependently decreased both basal and stimulated activation of p38 MAPK signaling in this drug resistant cell system. Decreased activation of p38 by RWJ67657 resulted in inhibition of the downstream p38 targets hsp27 and MAPKAPK. Diminished p38 signaling resulted in inhibition of p38-medated gene transcription. Furthermore, pharmacological inhibition of p38 by RWJ67657 decreased biological effects of p38, including ER-mediated gene expression and clonogenic survival in a dose-dependent manner. Animal studies revealed significantly decreased p38 signaling in vivo following exposure to RWJ67657. Treatment with the inhibitor markedly decreased phosphorylation of p38 in MDA-MB-361 tumors, leading to decreased transcription of both Fra-1 and progesterone receptor. Utilizing well-established xenograft tumor models, we demonstrated that RWJ67657 exhibits potent anti-tumor properties. Treatment with RWJ67657 markedly decreased tamoxifen resistant tumor growth, both in the presence and absence of estrogen. Taken together, our findings demonstrate the therapeutic potential of targeting the p38-MAPK signaling cascade in the treatment of endocrine resistant breast cancer.

3.
Cancer Biol Ther ; 13(11): 1026-33, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22825349

ABSTRACT

The p38 mitogen activated protein kinase pathway (MAPK) is known to promote cell survival, endocrine therapy resistance and hormone independent breast cancer cell proliferation. Therefore, we utilized the novel p38 inhibitor RWJ67657 to investigate the relevance of targeting this pathway in the ER (+) breast cancer cell line MCF-7. Our results show that RWJ67657 inhibits both basal and estrogen stimulated phosphorylation of p38α, resulting in decreased activation of the downstream p38α targets hsp27 and MAPAPK. Furthermore, inhibition of p38α by RWJ67657 blocks clonogenic survival of MCF-7 cells with little effect on non-cancerous breast epithelial cells. Even though p38α is known to phosphorylate ERα at residue within ER's hinge region at Thr311, resulting in increased ERα transcriptional activation, our results suggest RWJ67657 inhibits the p38α-induced activation of ER by targeting both the AF-1 and AF-2 activation domains within ERα. We further show that RWJ67657 decreases the transcriptional activity of the ER coactivators SRC-1, SRC-2 and SRC-3. Taken together, our results strongly suggest that in addition to phosphorylating Thr311 within ERα, p38α indirectly activates the ER by phosphorylation and stimulation of the known ERα coactivators, SRC-1, -2 and-3. Overall, our data underscore the therapeutic potential of targeting the p38 MAPK pathway in the treatment of ER (+) breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , src-Family Kinases/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Enzyme Activation , Estrogen Receptor alpha/genetics , Female , HEK293 Cells , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction , Transcriptional Activation , Transfection
4.
Neuro Oncol ; 13(7): 700-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21727209

ABSTRACT

Rapamycin, an anti-proliferative agent, is effective in the treatment of renal cell carcinoma and recurrent breast cancers. We proposed that this potent mammalian target of rapamycin inhibitor may be useful for the treatment of gliomas as well. We examined the cytotoxicity of rapamycin against a rodent glioma cell line, determined the toxicity of rapamycin when delivered intracranially, and investigated the efficacy of local delivery of rapamycin for the treatment of experimental malignant glioma in vivo. We also examined the dose-dependent efficacy of rapamycin and the effect when locally delivered rapamycin was combined with radiation therapy. Rapamycin was cytotoxic to 9L cells, causing 34% growth inhibition at a concentration of 0.01 µg/mL. No in vivo toxicity was observed when rapamycin was incorporated into biodegradable caprolactone-glycolide (35:65) polymer beads at 0.3%, 3%, and 30% loading doses and implanted intracranially. Three separate efficacy studies were performed to test the reproducibility of the effect of the rapamycin beads as well as the validity of this treatment approach. Animals treated with the highest dose of rapamycin beads tested (30%) consistently demonstrated significantly longer survival durations than the control and placebo groups. All dose-escalating rapamycin bead treatment groups (0.3%, 3% and 30%), treated both concurrently with tumor and in a delayed manner after tumor placement, experienced a significant increase in survival, compared with controls. Radiation therapy in addition to the simultaneous treatment with 30% rapamycin beads led to significantly longer survival duration than either therapy alone. These results suggest that the local delivery of rapamycin for the treatment of gliomas should be further investigated.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Glioma/drug therapy , Neoplasms, Experimental/drug therapy , Sirolimus/therapeutic use , Animals , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/radiation effects , Combined Modality Therapy , Glioma/pathology , Glioma/radiotherapy , Humans , Neoplasms, Experimental/pathology , Neoplasms, Experimental/radiotherapy , Polymers/chemistry , Radiotherapy , Rats , Rats, Inbred F344 , Survival Rate
5.
Cancer Res ; 71(2): 603-13, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21123450

ABSTRACT

Estrogen independence and progression to a metastatic phenotype are hallmarks of therapeutic resistance and mortality in breast cancer patients. Metastasis has been associated with chemokine signaling through the SDF-1-CXCR4 axis. Thus, the development of estrogen independence and endocrine therapy resistance in breast cancer patients may be driven by SDF-1-CXCR4 signaling. Here we report that CXCR4 overexpression is indeed correlated with worse prognosis and decreased patient survival irrespective of the status of the estrogen receptor (ER). Constitutive activation of CXCR4 in poorly metastatic MCF-7 cells led to enhanced tumor growth and metastases that could be reversed by CXCR4 inhibition. CXCR4 overexpression in MCF-7 cells promoted estrogen independence in vivo, whereas exogenous SDF-1 treatment negated the inhibitory effects of treatment with the anti-estrogen ICI 182,780 on CXCR4-mediated tumor growth. The effects of CXCR4 overexpression were correlated with SDF-1-mediated activation of downstream signaling via ERK1/2 and p38 MAPK (mitogen activated protein kinase) and with an enhancement of ER-mediated gene expression. Together, these results show that enhanced CXCR4 signaling is sufficient to drive ER-positive breast cancers to a metastatic and endocrine therapy-resistant phenotype via increased MAPK signaling. Our findings highlight CXCR4 signaling as a rational therapeutic target for the treatment of ER-positive, estrogen-independent breast carcinomas needing improved clinical management.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , Receptors, CXCR4/biosynthesis , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Humans , MAP Kinase Signaling System , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Receptors, CXCR4/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/biosynthesis
6.
J Biomed Biotechnol ; 2010: 597641, 2010.
Article in English | MEDLINE | ID: mdl-20625499

ABSTRACT

The long-term success of arthroplastic joints is dependent on the stabilization of the implant within the skeletal site. Movement of the arthroplastic implant within the bone can stimulate osteolysis, and therefore methods which promote rigid fixation or bone growth are expected to enhance implant stability and the long-term success of joint arthroplasty. In the present study, we used a simple bilateral bone defect model to analyze the osteogenic activity of three small-molecule drug implants via microcomputerized tomography (micro-CT) and histomorphometry. In this study, we show that local delivery of alendronate, but not lovastatin or omeprazole, led to significant new bone formation at the defect site. Since alendronate impedes osteoclast-development, it is theorized that alendronate treatment results in a net increase in bone formation by preventing osteoclast mediated remodeling of the newly formed bone and upregulating osteoblasts.


Subject(s)
Alendronate/pharmacology , Femur/drug effects , Femur/pathology , Lovastatin/pharmacology , Omeprazole/pharmacology , Osteogenesis/drug effects , Animals , Bone Density/drug effects , Disease Models, Animal , Femur/diagnostic imaging , Femur/surgery , Male , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Wound Healing/drug effects , X-Ray Microtomography
7.
Biomed Mater ; 4(1): 015001, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18981543

ABSTRACT

Post-surgical adhesion formation has numerous deleterious side effects in a wide variety of surgical settings. Physical barriers used together with laparoscopy were developed to reduce tissue trauma seen with open procedures. However, despite surgeons' meticulous techniques and the use of such barriers, adhesion formation remains a serious clinical problem, creating complications that cost the health care system over $1 billion annually. Our laboratories have combined a previously marketed drug, Tranilast, with a sodium carboxymethylcellulose (NaCMC) gel in a sustained release formulation using poly(p-dioxanone) (PDO) to provide a locally delivered medicated device that significantly reduces adhesions. This paper describes the preparation of the gel and the sustained release formulation, its key physical properties, and its sustained release kinetics. Pre-clinical data on inhibition of adhesion formation by the sustained release poly(p-dioxanone)/sodium carboxymethylcellulose/Tranilast drug enhanced device are also presented.


Subject(s)
Delayed-Action Preparations/chemistry , Postoperative Complications/prevention & control , Tissue Adhesions/prevention & control , ortho-Aminobenzoates/administration & dosage , ortho-Aminobenzoates/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Delayed-Action Preparations/administration & dosage , Diffusion , Drug Synergism , Humans , Rabbits , Rats
8.
Carcinogenesis ; 30(1): 106-13, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18791200

ABSTRACT

The activity of nuclear transcription factors is often regulated by specific kinase-signaling pathways. We have previously shown that the organochlorine pesticide dichlorodiphenyltrichloroethane (DDT) stimulates activator protein-1 activity through the p38 mitogen-activated protein kinase (MAPK). Here, we show that DDT and its metabolites also stimulate the transcriptional activity of cyclic adenosine monophosphate response element-binding protein and Elk1 and potentiate gene expression through cyclic adenosine monophosphate and hypoxia response elements. Because DDT stimulates gene expression through various transcription factors and hence multiple response elements, we hypothesized that p38 signaling targets a common shared transcriptional activator. Here, we demonstrate using both pharmacological and molecular techniques, the general coactivator p300 is phosphorylated and potentiated by the p38 MAPK signaling cascade. We further show that p38 directly phosphorylates p300 in its N-terminus. These results, together with our previous work, suggest that p38 stimulates downstream transcription factors in part by targeting the general coactivator p300.


Subject(s)
DDT/pharmacology , p300-CBP Transcription Factors/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Blotting, Western , Cell Line , Genes, Reporter , Humans , MAP Kinase Signaling System , Phosphorylation , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic , p300-CBP Transcription Factors/metabolism
9.
Diabetes Metab Syndr Obes ; 2: 91-100, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-21437122

ABSTRACT

In healthy tissue, a wound initiates an inflammatory response characterized by the presence of a hematoma, infiltration of inflammatory cells into the wound and, eventually, wound healing. In pathological conditions like diabetes mellitus, wound healing is impaired by the presence of chronic nonresolving inflammation. p38 mitogen-activated protein kinase (MAPK) inhibitors have demonstrated anti-inflammatory effects, primarily by inhibiting the expression of inflammatory cytokines and regulating cellular traffic into wounds. The db/db mouse model of type 2 diabetes was used to characterize the time course of expression of activated p38 during impaired wound healing. The p38α-selective inhibitor, SCIO-469, was applied topically and effects on p38 activation and on wound healing were evaluated. A topical dressing used clinically, Promogran™, was used as a comparator. In this study, we established that p38 is phosphorylated on Days 1 to 7 post-wounding in db/db mice. Further, we demonstrated that SCIO-469, at a dose of 10 µg/wound, had a positive effect on wound contraction, granulation tissue formation, and re-epithelialization, and also increased wound maturity during healing. These effects were similar to or greater than those observed with Promogran™. These results suggest a novel approach to prophylactic and therapeutic management of chronic wounds associated with diabetes or other conditions in which healing is impaired.

10.
Semin Reprod Med ; 26(4): 341-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18756411

ABSTRACT

Postsurgical adhesion formation has numerous deleterious side effects in a wide variety of surgical settings. Physical barriers used together with laparoscopy were developed in hopes of reducing the tissue trauma seen with open procedures and separating tissues during the critical time of healing to reduce adhesion formation. Despite meticulous techniques by surgeons and the availability of barriers, adhesion formation remains a serious problem, with more than $1 billion spent annually on complications arising from adhesions. Our laboratories have combined a previously marketed drug, Tranilast, with a gel to provide a locally delivered medicated device that can reduce adhesion formation. This article will review the role of Tranilast in the key pathways involved in adhesion formation.


Subject(s)
Peritoneal Diseases/prevention & control , Tissue Adhesions/prevention & control , ortho-Aminobenzoates/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Equipment and Supplies , Fibrosis/drug therapy , Fibrosis/prevention & control , Gynecologic Surgical Procedures/instrumentation , Gynecologic Surgical Procedures/methods , Humans , Peritoneal Diseases/metabolism , Peritoneum/drug effects , Peritoneum/pathology , Tissue Adhesions/metabolism , ortho-Aminobenzoates/administration & dosage
11.
Antimicrob Agents Chemother ; 51(12): 4324-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923491

ABSTRACT

We recently showed that the pyridinylimidazoles SB203580 and SB202190, drugs designed to block human p38 mitogen-activated protein kinase (MAPK) activation, also inhibited replication of the medically important intracellular parasite Toxoplasma gondii in cultured human fibroblasts through a direct effect on the parasite. We now show that additional pyridinylimidazole and imidazopyrimidine p38 MAPK inhibitors inhibit intracellular T. gondii replication in vitro and protect mice against fatal T. gondii infection. Mice surviving infection following treatment with p38 MAPK inhibitors were resistant to subsequent T. gondii challenge, demonstrating induction of protective immunity. Thus, drugs originally developed to block human p38 MAPK activation are useful for treating T. gondii infection without inducing significant immunosuppression. MAPK inhibitors combined with either of the approved anti-Toxoplasma drugs sulfadiazine and pyrimethamine resulted in improved survival among mice challenged with a fatal T. gondii inoculum. A MAPK inhibitor also treated mice infected with the Microsporidium parasite Encephalitozoon cuniculi, suggesting that MAPK inhibitors represent a novel class of agents that may have a broad spectrum of antiparasitic activity. Preliminary studies implicate a T. gondii MAPK homologue as the target of drug action, suggesting possibilities for more-selective agents.


Subject(s)
Antiprotozoal Agents/pharmacology , Encephalitozoonosis/drug therapy , Toxoplasmosis, Animal/drug therapy , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , CD8 Antigens/genetics , Dose-Response Relationship, Drug , Drug Design , Drug Synergism , Drug Therapy, Combination , Encephalitozoon cuniculi/drug effects , Encephalitozoonosis/prevention & control , Enzyme Inhibitors/pharmacology , Female , Humans , Imidazoles/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Pyridines/pharmacology , Time Factors , Toxoplasma/drug effects , Toxoplasmosis, Animal/genetics , Toxoplasmosis, Animal/prevention & control
12.
J Surg Res ; 141(2): 153-61, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17560610

ABSTRACT

BACKGROUND: Preclinical studies using the rabbit sidewall and double uterine horn models were used to assess time and dose response of tranilast delivered via subcutaneous pump, p.o., or as an intraperitoneal bolus in viscoelastic gels as well as an intraperitoneal biodegradable poly(p-dioxanone) fiber in reducing adhesions compared to vehicle controls. MATERIALS AND METHODS: New Zealand white rabbits underwent laparotomy followed by: 1) uterine horn abrasion and peripheral devascularization or 2) cecal abrasion and sidewall deperitonealization. Tranilast treatment using various vehicles and dosages was compared to vehicle alone versus no treatment. Animals were euthanized after 7 to 21 days. Adhesion formation was assessed by two independent observers. RESULTS: There were reductions in adhesion formation when drug was delivered topically, but oral drug alone was not effective. When tranilast was given preoperatively, oral drug added to the adhesion reduction of intraperitoneal administered drug. Tranilast in a viscoelastic carrier as well as in a biodegradable fiber was effective at reducing adhesions in the double uterine horn model. The slow release of tranilast from a biodegradable rod produced overall the best results. There were no safety issues. CONCLUSION: Tranilast was effective in reducing adhesions when given in a variety of vehicles in different rabbit models of adhesion formation. Overall, the sustained intraperitoneal delivery of tranilast from biodegradable fibers was the most suitable for clinical testing.


Subject(s)
Postoperative Complications/prevention & control , Tissue Adhesions/prevention & control , ortho-Aminobenzoates/administration & dosage , Animals , Drug Delivery Systems , Female , Laparotomy , Rabbits
13.
Bioorg Med Chem Lett ; 16(23): 6102-6, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16971122

ABSTRACT

Inhibition of the p38 map kinase pathway has been shown to be beneficial in the treatment of inflammatory diseases. The first class of potent p38 kinase inhibitors was the pyridinylimidazole compounds from SKB. Since then several pyridinylimidazole-based compounds have been shown to inhibit activated p38 kinase in vitro and in vivo. We have developed a novel series of pyridinylimidazole-based compounds, which potently inhibit the p38 pathway by binding to unactivated p38 kinase and only weakly inhibiting activated p38 kinase activity in vitro.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Esters/chemistry , Mice , Molecular Structure , Piperazine , Piperazines/chemistry , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Neurosci Methods ; 158(1): 22-9, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16797718

ABSTRACT

We report here a method for proteomics pattern discovery by utilizing a self-organizing map approach to analyze data obtained from a novel multiplex iTRAQ proteomics method. Through the application of this technique, we were able to delineate the early molecular events preceding dorsal root ganglia neurite outgrowth induced by either nerve growth factor (NGF) or an immunophilin ligand, JNJ460. Following pattern analysis we discovered that each neurotrophic agent promoted mostly distinct increases in protein expression with few overlapping patterns. In the NGF-treated group, proteins possessing "biosynthesis function" (p < 0.002) and "ribosome localization" (p < 0.0003) were increased, while proteins promoting "organogenesis" (p < 0.004) and related "signal transduction" (p < 0.008) functions were notably increased in the JNJ460-treated group. This study suggests that the properties of neurite outgrowth triggered by NGF and JNJ460 can be distinguished at the proteome level. Multiplexed proteomics analysis, along with pattern discovery bioinformatics tools, has the capability to differentiate subtle neuroproteomics patterns.


Subject(s)
Cell Differentiation/physiology , Neurites/metabolism , Neurons/physiology , Proteomics/methods , Animals , Blotting, Western/methods , Cell Differentiation/drug effects , Computational Biology/methods , Electrophoresis, Gel, Two-Dimensional/methods , Ganglia, Spinal/cytology , Mice , Mice, Inbred C57BL , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Tandem Mass Spectrometry/methods
15.
Blood ; 106(6): 2059-68, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15928037

ABSTRACT

The human immunodeficiency virus (HIV) has been reported to target noninfected CD4 and CD8 cells for destruction. This effect is manifested in part through up-regulation of the death receptor Fas ligand (FasL) by HIV-1 negative factor (Nef), leading to bystander damage. However, the signal transduction and transcriptional regulation of this process remains elusive. Here, we provide evidence that p38 mitogen-activated protein kinase (MAPK) is required for this process. Loss-of-function experiments through dominant-negative p38 isoform, p38 siRNA, and chemical inhibitors of p38 activation suggest that p38 is necessary for Nef-induced activator protein-1 (AP-1) activation, as inhibition leads to an attenuation of AP-1-dependent transcription. Furthermore, mutagenesis of the FasL promoter reveals that its AP-1 enhancer element is required for Nef-mediated transcriptional activation. Therefore, a linear pathway for Nef-induced FasL expression that encompasses p38 and AP-1 has been elucidated. Furthermore, chemical inhibition of the p38 pathway attenuates HIV-1-mediated bystander killing of CD8 cells in vitro.


Subject(s)
Bystander Effect , CD8-Positive T-Lymphocytes/pathology , Gene Products, nef/physiology , HIV-1/pathogenicity , Membrane Glycoproteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Death , Cells, Cultured , Enhancer Elements, Genetic , Fas Ligand Protein , Gene Expression Regulation , Humans , Transcription Factor AP-1 , Transcriptional Activation , nef Gene Products, Human Immunodeficiency Virus
16.
J Biol Chem ; 279(27): 27845-8, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15133031

ABSTRACT

FK506-binding protein 52 (FKBP52) is an immunophilin that possesses peptidylprolyl cis/trans-isomerase (PPIase) activity and is a component of a subclass of steroid hormone receptor complexes. Several recent studies indicate that immunophilins can regulate neuronal survival and nerve regeneration although the molecular mechanisms are poorly understood. To investigate the function of FKBP52 in the nervous system, we employed a yeast two-hybrid strategy using the PPIase domain (domain I) as bait to screen a neonatal rat dorsal root ganglia cDNA expression library. We identified an interaction between FKBP52 domain I and Atox1, a copper-binding metallochaperone. Atox1 interacts with Menkes disease protein and Wilson disease protein (WD) and functions in copper efflux. The interaction between FKBP52 and Atox1 was observed in both glutathione S-transferase pull-down experiments and when proteins were ectopically expressed in human embryonic kidney (HEK) 293T cells and was sensitive to FK506. Interestingly, the FKBP52/Atox1 interaction was enhanced when HEK 293T cells were cultured in copper-supplemented medium and decreased in the presence of the copper chelator, bathocuproine disulfate, suggesting that the interaction is regulated in part by intracellular copper. Overexpression of FKBP52 increased rapid copper efflux in (64)Cu-loaded cells, as did the overexpression of WD transporter. Taken together, our present findings suggest that FKBP52 is a component of the copper efflux machinery, and in so, may also promote neuroprotection from copper toxicity.


Subject(s)
Copper/metabolism , Immunophilins/metabolism , Tacrolimus Binding Proteins/physiology , Animals , Biological Transport , Blotting, Western , Calcium/metabolism , Cell Line , Cell Survival , Chelating Agents/pharmacology , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Gene Library , Genetic Vectors , Glutathione Transferase/metabolism , Humans , Kinetics , Models, Genetic , Neurons/metabolism , Phenanthrolines/pharmacology , Precipitin Tests , Protein Structure, Tertiary , Rats , Time Factors , Two-Hybrid System Techniques
17.
AIDS ; 18(5): 739-48, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-15075508

ABSTRACT

OBJECTIVE: To analyze a novel compound, which inhibits serine-threonine protein kinase p38, for its possible bioactivity against HIV-1 infection. METHODS: Proteins involved in cellular signal transduction pathways represent a novel class of host therapeutic targets for infectious diseases. In this regard the serine/threonine kinase p38 MAPK, a member of the mitogen-activated protein (MAP) kinase superfamily of signal transduction molecules may play an important role in HIV-1 infection. We analyzed the ability of this compound (RWJ67657) to inhibit HIV replication in primary T cells and monocytes. Cellular expression of phospho-p38MAPK was studied by Western blot analysis. Blockade of HIV infection induced apoptosis was measured by Annexin V staining. RESULTS: p38 inhibitor RWJ67657 was effective in inhibiting HIV-1 replication in both T-cell and monocyte cell lines, irrespective of the coreceptor used by the virus for entry into the cell. Importantly, both reverse transcriptase and protease resistant escape mutant viruses were effectively suppressed by RWJ67657. In addition, the tested compounds block HIV-induced T-cell apoptosis, a critical means of T-cell depletion linked to AIDS progression. CONCLUSION: Several steps in the HIV-1 virus life cycle appear to depend on cellular activation, including activation of the p38 pathway. Without activation virus replication is thought to be blocked due to incomplete reverse transcription and a lack of proviral DNA integration. The data collectively illustrate that inhibition of the p38 pathway can affect HIV-1 replication. Interruption of HIV infection by p38 inhibitors underscores the value of exploring antiviral drugs that target host cellular proteins.


Subject(s)
HIV-1/physiology , Imidazoles/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyridines/pharmacology , T-Lymphocytes/virology , Virus Replication/drug effects , Analysis of Variance , Annexin A5/analysis , Apoptosis , Biomarkers/analysis , Blotting, Western/methods , Cell Line , Drug Resistance, Viral , HIV-1/drug effects , Humans , Jurkat Cells , Mitogen-Activated Protein Kinases/metabolism , Monocytes/enzymology , Monocytes/pathology , Monocytes/virology , Phosphorylation , T-Lymphocytes/enzymology , T-Lymphocytes/pathology , U937 Cells , p38 Mitogen-Activated Protein Kinases
18.
Bioorg Med Chem Lett ; 13(3): 347-50, 2003 Feb 10.
Article in English | MEDLINE | ID: mdl-12565927

ABSTRACT

The MAP kinase p38 is implicated in the release of the pro-inflammatory cytokines TNF-alpha and IL-1 beta. Inhibition of cytokine release may be a useful treatment for inflammatory conditions such as rheumatoid arthritis and Crohn's disease. A novel series of imidazopyrimidines have been discovered that potently inhibit p38 and suppress the production of TNF-alpha in vivo.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Catalysis , Indicators and Reagents , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases
19.
Biochem J ; 363(Pt 1): 175-82, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11903060

ABSTRACT

T-cell activation requires signals from both the T-cell receptor (TcR) and other co-stimulatory molecules such as CD28. TcR- and CD28-mediated signals are integrated during T-cell activation resulting in the expression of cytokine genes such as interleukin-2 (IL-2). An enhancer element (CD28RE) of the IL-2 gene specifically responsive to CD28 signals has been previously identified and characterized. This response element and an adjacent Activated Protein-1 (nuclear factor-interleukin-2B) site together (RE/AP1) were shown to complex with c-rel, AP-1 and other factors. However, details of the signal transduction pathways leading from CD28 to the composite response element remain poorly understood. We present data showing that overexpression of the serine threonine kinase, mitogen-activated protein kinase/extracellular-signal-regulated kinase kinase kinase-1 (MEKK1), but not nuclear factor-kappa B inducing kinase, or MAP kinase/ERK kinase-1 (MEK1), can significantly increase the level of CD28RE/AP1-driven luciferase (Luc) reporter gene expression in Jurkat E6-1 cells. A MEKK1 dominant negative mutant blocked such activation induced by stimulation with Raji B cells and the superantigen staphylococcus enterotoxin E (SEE), as well as via CD3/CD28. Mutations in either site of the RE/AP1 element abolished MEKK1-induced Luc expression. Calcineurin inhibitors, CsA and FK520, or inhibitors of p38 kinase (SB 203580), or MEK1 (PD 098059), did not affect MEKK1-induced reporter activation. These results directly implicate MEKK1 in the CD28 signalling pathway that activates the CD28 response element. Co-expression of the lymphocyte-oriented kinase (LOK) kinase attenuated Raji/SEE-induced IL-2 production in Jurkat cells, as well as MEKK1 and Raji/SEE-induced reporter gene activation. These data suggest that MEKK1 and LOK may have opposing roles in regulating the CD28RE/AP1 element.


Subject(s)
CD28 Antigens/biosynthesis , MAP Kinase Kinase Kinase 1 , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , T-Lymphocytes/enzymology , CD28 Antigens/metabolism , Calcineurin/metabolism , Cell Line , DNA/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Enhancer Elements, Genetic , Enzyme Activation , Genes, Dominant , Genes, Reporter , Humans , Interleukin-2/metabolism , Jurkat Cells , Luciferases/metabolism , Lymphocyte Activation , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Mutation , NF-kappa B/metabolism , Plasmids/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...