Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
Basic Clin Androl ; 28: 14, 2018.
Article in English | MEDLINE | ID: mdl-30473791

ABSTRACT

BACKGROUND: Bacterial infection and inflammation of the testis impairs fertility, yet an understanding of inflammatory responses of the testis is incomplete. We are interested in identifying gene pathways involved in the detection and clearance of infectious microbes in the male reproductive tract. In previous studies in our lab focused on hypoxia-responsive genes of the testis, preliminary experiments suggested that genes classically categorized as hypoxia genes are also activated during antimicrobial responses. The purpose of this study was to identify hypoxia and inflammatory gene pathways that contribute to antimicrobial protection of the testis and to consider possible cross-talk and interactions between these pathways. Inflammation was induced in Sprague-Dawley rats using P. aeruginosa or E. coli lipopolysaccharide (LPS). Levels of hypoxia-inducible factor-1 (HIF-1α) protein and nuclear factor kappa B (NF-κB) were measured, and hypoxia and inflammatory gene expression patterns in testis were analyzed by gene expression profiling using real-time quantitative PCR arrays. RESULTS: In LPS-treated rats, HIF-1α protein increased with no change in Hif-1α mRNA. Western Blot analysis also demonstrated no change in NF-κB and inhibitory NFKB alpha (IκBα) protein levels following LPS treatment. Five hypoxia pathway genes (Angptl4, Egr1, Ier3, Pai1, and Glut1), and 11 inflammatory pathway genes (Ccl12, Cc13, Cd14, Cxcl10, Icam1, Il10, Il1b, Il6, Nfkbia, Tlr2, Tnf) up-regulated after 3 h of inflammation. Angptl4, Ccl12, Cc13, Cd14, Egr1, Nfkbia, Tlr2, and Tnf remained elevated at 6 h. Six genes were up-regulated at 6 h only (Bhlhe40, C3, Jak2, Nlrp3, Slc11a1, Tlr1). One gene (Tlr5) was down-regulated after 3 h and no genes at 6 h. Electrophoretic mobility shift assay results suggest a decrease in NF-κB binding activity following LPS treatment. CONCLUSIONS: Testicular HIF-1α is up-regulated following LPS-induced inflammation. In contrast to other tissues, in which HIF-1α is up-regulated through transcriptional activation via NF-κB, we conclude that HIF-1α in the testis is not up-regulated through an increase in Hif-1α mRNA or through NF-κB-dependent mechanisms. Hypoxia pathway genes and genes involved in Toll-like receptor (TLR) and cytokine-mediated signaling comprise major functional categories of up-regulated genes, demonstrating that both hypoxia and classic inflammatory pathways are involved in inflammatory responses of the testis.


CONTEXTE: L'infection et l'inflammation bactériennes du testicule altèrent la fertilité; cependant la compréhension des réponses inflammatoires du testicule est. encore incomplète. Nous nous sommes intéressés à l'identification des voies des gènes impliqués dans la détection et l'élimination des microbes infectieux dans l'appareil reproductif masculin. Dans de précédentes études menées dans notre laboratoire, et centrées sur des gènes sensibles à l'hypoxie, les expérimentations préliminaires suggéraient que les gènes classiquement catégorisés comme gènes de l'hypoxie étaient aussi activés au cours des réponses antimicrobiennes. Le but de la présente étude était d'identifier les voies des gènes qui contribuaient à la protection antimicrobienne du testicule et d'examiner de potentiels intermodulations et interactions entre ces voies.L'inflammation a été induite chez des rats Sprague-Dawley en utilisant des lypopolysaccharides (LPS) de P. aeruginosaet d'E. coli. Les taux de protéine du facteur-1 inductible par l'hypoxie (HIF1- α) et du facteur nucléaire kappa B (NF- kB) ont été mesurés; les profils d'expression des gènes de l'hypoxie et de l'inflammation dans le testicule ont été analysés par profilage de l'expression génique par PCR quantitative en temps réel. RÉSULTATS: Chez les rats traités par LPS, la protéine HIF-1 α a augmenté sans modification de Hif-1αmRNA. L'analyse par Western Blot a aussi montré l'absence de modifications des taux de NF-kB et de la protéine inhibitrice NFKB alpha (IkB α) après traitement. Cinq gènes de la voie hypoxie (Angptl4, Egr1, Ier3, Pai1,et Glut1), et 11 gènes de la voie inflammatoire (Ccl12, Cc13, Cd14, Cxcl10, Icam1, Il10, Il1b, Il6, Egr1, Nfkbia, Tlr2, et Tnf) ont été régulés à la hausse après 3 heures d'inflammation. Angptl4, Ccl12, Cc13, Cd14, Egr1, Nfkbia, Tlr2, et Tnfsont restés élevés à 6 heures. Six gènes n'ont ont été régulés à la hausse qu'à 6 heures (Bhlhe40, C3, Jak2, Nlrp3, Slc11a1, Tlr1). Un gène (Tlr5) a été régulé à la baisse après 3 heures et aucun gène à 6 heures. Les résultats du test de décalage de la mobilité électrophorétique suggèrent une baisse de l'activité de liaison de NF- kB après traitement par LPS. CONCLUSIONS: HIF-1α testiculaire est. régulé à la hausse après inflammation induite par LPS. Contrairement à d'autres tissus, dans lesquels HIF-1α est. régulé à la hausse par activation transcriptionnelle via NF- kB, nous concluons que HIF-1α dans le testicule n'est. pas régulé à la hausse par une augmentation de Hif-1 αmRNA ou par des mécanismes NF-kB-dépendants. Les gènes de la voie hypoxie et les gènes impliqués dans le récepteur Toll-like (TLR) et dans la signalisation médiée par les cytokines comprennent des catégories fonctionnelles majeures de gènes régulés à la hausse, ce qui démontre qu'à la fois les voies de l'hypoxie et les voies classiques de l'inflammation sont impliquées dans les réponses inflammatoires du testicule.

2.
Am J Reprod Immunol ; 77(6)2017 06.
Article in English | MEDLINE | ID: mdl-28328045

ABSTRACT

PROBLEM: Little is known about how infection and the response to inflammation affect the microRNA (miRNA) profile of the male reproductive tract. We hypothesized that expression of inflammatory-related miRNAs would be altered following immune activation of rat testis. METHOD OF STUDY: Testis total RNA was purified from Sprague-Dawley rats 3 or 6 hours after receiving a 5 mg/kg intraperitoneal injection of bacterial lipopolysaccharide (LPS) and examined by qPCR using an 84-panel miRNA array. RESULTS: Five inflammatory-related miRNAs showed a greater than twofold downregulation (P<.05) in the 3-hour group (rno-let-7f-5p, rno-miR-200c-3p, rno-miR-23a-3p, rno-miR-23b-3p, rno-miR-98-5p) and five from the 6-hour group (rno-miR-17-5p, rno-miR-19a-3p, rno-miR-34a-5p, rno-miR-34c-5p, rno-miR-449a-5p). CONCLUSION: Review of the literature has revealed that these miRNAs also play important roles in the maintenance of fertility, formation and elimination of cancer, and development of the male reproductive tract. Further study will lead to a greater understanding of male reproductive immunology and related health issues.


Subject(s)
Inflammation/genetics , MicroRNAs/genetics , Testis/immunology , Animals , Down-Regulation , Inflammation/immunology , Lipopolysaccharides/pharmacology , Male , MicroRNAs/immunology , Rats, Sprague-Dawley
3.
Chem Biol ; 21(8): 955-66, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25065531

ABSTRACT

The effect of acanthoic acid analogs on the response to proinflammatory challenge was investigated. Some pimarane diterpenes are known activators of the LXRαß nuclear receptors, but we show here that they also exert a rapid, potent, and selective activation of the p110γ and p110δ subunits of PI3K. Combination of these effects results in an important attenuation of the global transcriptional response to LPS in macrophages. PI3K/Akt activation leads to inhibition of the LPS-dependent stimulation of IKK/NF-κB and p38 and ERK MAPKs. Macrophages from LXRαß-deficient mice exhibited an inhibition of these pathways similar to the corresponding wild-type cells. Silencing or inhibition of p110γ/δ suppressed the effect of these diterpenes (DTPs) on IKK/NF-κB and MAPKs signaling. Taken together, these data show a multitarget anti-inflammatory mechanism by these DTPs including a selective activation of PI3K isoenzymes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Diterpenes/pharmacology , NF-kappa B/antagonists & inhibitors , Protein Subunits/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Diterpenes/chemistry , Dose-Response Relationship, Drug , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Liver X Receptors , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Conformation , NF-kappa B/metabolism , NIH 3T3 Cells , Orphan Nuclear Receptors/deficiency , Orphan Nuclear Receptors/metabolism , Protein Subunits/chemistry , Structure-Activity Relationship
4.
Reprod Biol Endocrinol ; 10: 104, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23216940

ABSTRACT

BACKGROUND: Spermatic cord torsion can lead to testis ischemia (I) and subsequent ischemia-reperfusion (I/R) causing germ cell-specific apoptosis. Previously, we demonstrated that the hypoxia-inducible factor-1 (HIF-1) transcription factor, a key regulator of physiological responses to hypoxia, is abundant in Leydig cells in normoxic and ischemic testes. We hypothesize that testicular HIF-1 activates the expression of antiapoptotic target genes to protect Leydig cells from apoptosis. In silico analysis of testis genes containing a consensus hypoxia response element (HRE, 5'-RCGTG-3') identified myeloid cell leukemia-1 (Mcl-1) as a potential HIF-1 target gene. The purpose of this study was to determine whether HIF-1 shows DNA-binding activity in normoxic and ischemic testes and whether Mcl-1 is a target gene of testicular HIF-1. METHODS: The testicular HIF-1 DNA-binding capacity was analyzed in vitro using a quantitative enzyme-linked immunosorbent assay (ELISA) and electrophoretic mobility shift assays (EMSA). MCL-1 protein expression was evaluated by immunoblot analysis and immunohistochemistry. The binding of testicular HIF-1 to the Mcl-1 gene was examined via chromatin immunoprecipitation (ChIP) analysis. RESULTS: The ELISA and EMSA assays demonstrated that testicular HIF-1 from normoxic and ischemic testes binds DNA equally strongly, suggesting physiological roles for HIF-1 in the normoxic testis, unlike most tissues in which HIF-1 is degraded under normoxic conditions and is only activated by hypoxia. MCL-1 protein was determined to be abundant in both normoxic and ischemic testes and expressed in Leydig cells. In a pattern identical to that of HIF-1 expression, the steady-state levels of MCL-1 were not significantly affected by I or I/R and MCL-1 co-localized with HIF-1α in Leydig cells. Chromatin immunoprecipitation (ChIP) analysis using a HIF-1 antibody revealed sequences enriched for the Mcl-1 promoter. CONCLUSIONS: The results demonstrated that, unlike what is observed in most tissues, HIF-1 displays DNA-binding activity in both normoxic and ischemic testes, and Mcl-1 may be a key target gene of testicular HIF-1 with potential roles in the antiapoptotic protection of Leydig cells.


Subject(s)
Hypoxia-Inducible Factor 1/analysis , Hypoxia-Inducible Factor 1/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Testis/chemistry , Animals , Apoptosis , Cell Hypoxia , Chromatin Immunoprecipitation , DNA/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Immunoblotting , Immunohistochemistry , Ischemia/metabolism , Leydig Cells/chemistry , Male , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/analysis , Rats , Rats, Sprague-Dawley , Testis/blood supply
5.
Planta Med ; 78(2): 131-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22034066

ABSTRACT

Four ß-lactone- γ-lactam proteasome inhibitors of natural origin were tested for their trypanocidal activities in vitro using culture-adapted bloodstream forms of Trypanosoma brucei. All four compounds displayed activities in the nanomolar range. The most trypanocidal compounds with 50% growth inhibition (GI(50)) values of around 3 nM were the bromine and iodine analogues of salinosporamide A, a potent proteasome inhibitor produced by the marine actinomycete Salinispora tropica. In general, trypanosomes were more susceptible to the compounds than were human HL-60 cells. The data support the potential of ß-lactone- γ-lactam proteasome inhibitors for rational anti-trypanosomal drug development.


Subject(s)
Biological Products/pharmacology , Enzyme Inhibitors/pharmacology , Lactams/pharmacology , Lactones/pharmacology , Proteasome Inhibitors , Pyrroles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects
6.
Invest New Drugs ; 30(6): 2303-17, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22080430

ABSTRACT

PURPOSE: Combining proteasome and histone deacetylase (HDAC) inhibition has been seen to provide synergistic anti-tumor activity, with complementary effects on a number of signaling pathways. The novel bi-cyclic structure of marizomib with its unique proteasome inhibition, toxicology and efficacy profiles, suggested utility in combining it with an HDAC inhibitor such as vorinostat. Thus, in this study in vitro studies assessed the potential utility of combining marizomib and vorinostat, followed by a clinical trial with the objectives of assessing the recommended phase 2 dose (RP2D), pharmacokinetics (PK), pharmacodynamics (PD), safety and preliminary anti-tumor activity of the combination in patients. EXPERIMENTAL DESIGN: Combinations of marizomib and vorinostat were assessed in vitro. Subsequently, in a Phase 1 clinical trial patients with melanoma, pancreatic carcinoma or Non-small Cell Lung Cancer (NSCLC) were given escalating doses of weekly marizomib in combination with vorinostat 300 mg daily for 16 days in 28 day cycles. In addition to standard safety studies, proteasome inhibition and pharmacokinetics were assayed. RESULTS: Marked synergy of marizomib and vorinostat was seen in tumor cell lines derived from patients with NSCLC, melanoma and pancreatic carcinoma. In the clinical trial, 22 patients were enrolled. Increased toxicity was not seen with the combination. Co-administration did not appear to affect the PK or PD of either drug in comparison to historical data. Although no responses were demonstrated using RECIST criteria, 61% of evaluable patients demonstrated stable disease with 39% having decreases in tumor measurements. CONCLUSIONS: Treatment of multiple tumor cell lines with marizomib and vorinostat resulted in a highly synergistic antitumor activity. The combination of full dose marizomib with vorinostat is tolerable in patients with safety findings consistent with either drug alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Pancreatic Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis/drug effects , Carcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Combinations , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Lactones/administration & dosage , Lactones/blood , Lactones/pharmacokinetics , Lung Neoplasms/metabolism , Male , Melanoma/metabolism , Middle Aged , Pancreatic Neoplasms/metabolism , Proteasome Inhibitors/administration & dosage , Proteasome Inhibitors/blood , Proteasome Inhibitors/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/blood , Pyrroles/pharmacokinetics , Vorinostat
7.
Chem Biol Interact ; 194(1): 58-68, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21864512

ABSTRACT

Marizomib (NPI-0052) is a naturally derived irreversible proteasome inhibitor that potently induces apoptosis via a caspase-8 and ROS-dependent mechanism in leukemia cells. We aim to understand the relationship between the irreversible inhibition of the proteasome and induction of cell death in leukemia cells by using analogs of marizomib that display reversible and irreversible properties. We highlight the importance of sustained inhibition of at least two proteasome activities as being key permissive events for the induction of the apoptotic process in leukemia cells. These data provide the basis for the development of new approaches to generate more effective anti-proteasome therapies.


Subject(s)
Apoptosis/drug effects , Lactones/pharmacology , Protease Inhibitors/pharmacology , Pyrroles/pharmacology , Caspase 8/metabolism , Humans , Lactones/chemistry , Leukemia/drug therapy , Leukemia/metabolism , Leukemia/pathology , Oxidative Stress/drug effects , Protease Inhibitors/chemistry , Pyrroles/chemistry , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
8.
Blood ; 117(21): 5692-700, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21454451

ABSTRACT

Previous studies have established a role of vascular-disrupting agents as anti- cancer agents. Plinabulin is a novel vascular-disrupting agent that exhibits potent interruption of tumor blood flow because of the disruption of tumor vascular endothelial cells, resulting in tumor necrosis. In addition, plinabulin exerts a direct action on tumor cells, resulting in apoptosis. In the present study, we examined the anti-multiple myeloma (MM) activity of plinabulin. We show that low concentrations of plinabulin exhibit a potent antiangiogenic action on vascular endothelial cells. Importantly, plinabulin also induces apoptotic cell death in MM cell lines and tumor cells from patients with MM, associated with mitotic growth arrest. Plinabulin-induced apoptosis is mediated through activation of caspase-3, caspase-8, caspase-9, and poly(ADP-ribose) polymerase cleavage. Moreover, plinabulin triggered phosphorylation of stress response protein JNK, as a primary target, whereas blockade of JNK with a biochemical inhibitor or small interfering RNA strategy abrogated plinabulin-induced mitotic block or MM cell death. Finally, in vivo studies show that plinabulin was well tolerated and significantly inhibited tumor growth and prolonged survival in a human MM.1S plasmacytoma murine xenograft model. Our study therefore provides the rationale for clinical evaluation of plinabulin to improve patient outcome in MM.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 8/metabolism , Multiple Myeloma/blood supply , Multiple Myeloma/drug therapy , Neovascularization, Pathologic/prevention & control , Piperazines/pharmacology , Animals , Blotting, Western , Cell Proliferation/drug effects , Diketopiperazines , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
J Pharmacol Exp Ther ; 337(2): 479-86, 2011 May.
Article in English | MEDLINE | ID: mdl-21303921

ABSTRACT

The present study was undertaken to compare the cellular transport characteristics of [(3)H]NPI-0052 (1R,4R,5S)-4-(2-chloroethyl)-1-((S)-((S)-cyclohex-2-enyl)(hydroxy)methyl)-5-methyl-6-oxa-2-azabicyclo[3.2.0]heptane-3,7-dione (marizomib; salinosporamide A) and [(3)H]NPI-0047 (1R,4R, 5S)-1-((S)-((S)-cyclohex-2-enyl)(hydroxy)methyl)-4-ethyl-5-methyl-6-oxa-2-azabicyclo[3.2.0]heptane-3,7-dione in RPMI 8226 multiple myeloma and PC-3 prostate adenocarcinoma cells to determine whether these properties explain differences in the cytotoxic potencies of these chemical analogs. The results indicate that marizomib, which possesses a chemical-leaving group, is more cytotoxic to both cell lines and inhibits proteasome activity more completely at lower concentrations than NPI-0047, a nonleaving-group analog. Moreover, it was found that both compounds accumulate in these cells by simple diffusion and the same carrier-mediated transport system. Although the rate of uptake is similar, the cellular efflux, which does not seem to be mediated by a major ATP-binding cassette (ABC)-efflux transporter, is more rapid for NPI-0047 than for marizomib. Experiments revealed that the irreversible binding of marizomib to the proteasome is responsible for its slower efflux, longer duration of action, and greater cytotoxicity compared with NPI-0047. The discovery that major ABC transporters of the multidrug resistance-associated protein family do not seem to be involved in the accumulation or removal of these agents suggests they may not be affected by multidrug resistance mechanisms during prolonged administration.


Subject(s)
Antineoplastic Agents/pharmacology , Lactones/pharmacology , Proteasome Endopeptidase Complex/drug effects , Pyrroles/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lactams/metabolism
10.
Am J Pathol ; 177(2): 735-43, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20595632

ABSTRACT

NOD-like receptors (NLRs) and caspase-1 are critical components of innate immunity, yet their over-activation has been linked to a long list of microbial and inflammatory diseases, including anthrax. The Bacillus anthracis lethal toxin (LT) has been shown to activate the NLR Nalp1b and caspase-1 and to induce many symptoms of the anthrax disease in susceptible murine strains. In this study we tested whether it is possible to prevent LT-mediated disease by pharmacological inhibition of caspase-1. We found that caspase-1 and proteasome inhibitors blocked LT-mediated caspase-1 activation and cytolysis of LT-sensitive (Fischer and Brown-Norway) rat macrophages. The proteasome inhibitor NPI-0052 also prevented disease progression and death in susceptible Fischer rats and increased survival in BALB/c mice after LT challenge. In addition, NPI-0052 blocked rapid disease progression and death in susceptible Fischer rats and BALB/c mice challenged with LT. In contrast, Lewis rats, which harbor LT-resistant macrophages, showed no signs of caspase-1 activation after LT injection and did not exhibit rapid disease progression. Taken together, our findings indicate that caspase-1 activation is critical for rapid disease progression in rodents challenged with LT. Our studies indicate that pharmacological inhibition of NLR signaling and caspase-1 can be used to treat inflammatory diseases.


Subject(s)
Anthrax/metabolism , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Caspase 1/metabolism , Proteasome Inhibitors , Animals , Bacillus anthracis/pathogenicity , Caspase Inhibitors , Cell Death , Cells, Cultured , Enzyme Activation , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Inbred Strains
11.
Br J Haematol ; 149(4): 550-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20331453

ABSTRACT

Our previous study showed that the novel proteasome inhibitor NPI-0052 induces apoptosis in multiple myeloma (MM) cells resistant to conventional and bortezomib (Velcade, Takeda, Boston, MA, USA) therapies. In vivo studies using human MM-xenografts demonstrated that NPI-0052 is well tolerated, prolongs survival, and reduces tumour recurrence. These preclinical studies provided the basis for an ongoing phase-1 clinical trial of NPI-0052 in relapsed/refractory MM patients. Here we performed pharmacodynamic (PD) studies of NPI-0052 using human MM xenograft murine model. Our results showed that NPI-0052: (i) rapidly left the vascular compartment in an active form after intravenous (i.v.) administration, (ii) inhibited 20S proteasome chymotrypsin-like (CT-L, beta5), trypsin-like (T-L, beta2), and caspase-like (C-L, beta1) activities in extra-vascular tumours, packed whole blood (PWB), lung, liver, spleen, and kidney, but not brain and (iii) triggered a more sustained (>24 h) proteasome inhibition in tumours and PWB than in other organs (<24 h). Tissue distribution analysis of radiolabeled compound (3H-NPI-0052) in mice demonstrated that NPI-0052 left the vascular space and entered organs as the parent compound. Importantly, treatment of MM.1S-bearing mice with NPI-0052 showed reduced tumour growth without significant toxicity, which was associated with prolonged inhibition of proteasome activity in tumours and PWB but not normal tissues.


Subject(s)
Antineoplastic Agents/therapeutic use , Lactones/therapeutic use , Plasmacytoma/drug therapy , Pyrroles/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Kidney/metabolism , Lactones/pharmacokinetics , Lactones/pharmacology , Male , Mice , Plasmacytoma/metabolism , Plasmacytoma/pathology , Proteasome Inhibitors , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Blood ; 115(4): 834-45, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19965674

ABSTRACT

Our recent study demonstrated that a novel proteasome inhibitor NPI-0052 is distinct from bortezomib (Velcade) and, importantly, triggers apoptosis in multiple myeloma (MM) cells resistant to bortezomib. Here we demonstrate that combining NPI-0052 and lenalidomide (Revlimid) induces synergistic anti-MM activity in vitro using MM-cell lines or patient MM cells. NPI-0052 plus lenalidomide-induced apoptosis is associated with (1) activation of caspase-8, caspase-9, caspase-12, caspase-3, and poly(ADP) ribose polymerase; (2) activation of BH-3 protein BIM; (3) translocation of BIM to endoplasmic reticulum; (4) inhibition of migration of MM cells and angiogenesis; and (5) suppression of chymotrypsin-like, caspase-like, and trypsin-like proteasome activities. Importantly, blockade of BIM using siRNA significantly abrogates NPI-0052 plus lenalidomide-induced apoptosis. Furthermore, studies using biochemical inhibitors of caspase-8 versus caspase-9 demonstrate that NPI-0052 plus lenalidomide-triggered apoptosis is primarily dependent on caspase-8 signaling. In animal tumor model studies, low-dose combination of NPI-0052 and lenalidomide is well tolerated, significantly inhibits tumor growth, and prolongs survival. Taken together, our study provides the preclinical rationale for clinical protocols evaluating lenalidomide together with NPI-0052 to improve patient outcome in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Lactones/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Pyrroles/pharmacology , Thalidomide/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Drug Synergism , Humans , In Vitro Techniques , Lenalidomide , Mice , Mice, SCID , Proteasome Inhibitors , Thalidomide/pharmacology , Xenograft Model Antitumor Assays
13.
Bioorg Med Chem ; 17(6): 2175-80, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19022674

ABSTRACT

The discovery of the anticancer agent salinosporamide A (NPI-0052) resulted from the exploration of new marine environments and a commitment to the potential of the ocean to yield new natural products for drug discovery and development. Driving the success of this process was the linkage of academic research together with the ability and commitment of industry to undertake drug development and provide the resources and expertise to advance the entry of salinosporamide A (NPI-0052) into human clinical trials. This paper offers a chronicle of the important events that facilitated the rapid clinical development of this exciting molecule.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery , Lactones/chemistry , Pyrroles/chemistry , Drugs, Investigational , Molecular Structure
14.
Am J Reprod Immunol ; 60(6): 541-55, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19032616

ABSTRACT

PROBLEM: Toll-like receptors (TLRs) are a family of innate immunity receptors that are essential for detecting invading pathogens. Objectives of this study were to identify epididymal cell types expressing TLRs and to determine if TLRs are present on spermatozoa. METHOD OF STUDY: Immunohistochemical analysis of paraffin-embedded sections from regions of the rat epididymis was used to identify epididymal cell types expressing TLRs. Immunoblot analysis and immunofluorescence were used to detect TLRs on spermatozoa. RESULTS: TLRs 1-9 and 11 are abundantly expressed by the epididymal epithelium in most regions of the duct with the exception of clear cells of the cauda which do not express TLRs 5-7 or 11. TLRs were detected on epididymal spermatozoa and several TLRs showed regional distributions patterns suggesting modification during epididymal transit. CONCLUSION: The abundance of TLR family members in the epididymal epithelium and on spermatozoa provides strong evidence that TLRs play important roles in innate immunity of the male reproductive tract.


Subject(s)
Epididymis/cytology , Epididymis/metabolism , Epithelial Cells/metabolism , Spermatozoa/metabolism , Toll-Like Receptors/metabolism , Animals , Immunohistochemistry , Male , Organ Specificity/immunology , Rats , Rats, Sprague-Dawley
15.
J Med Chem ; 51(21): 6711-24, 2008 Nov 13.
Article in English | MEDLINE | ID: mdl-18939815

ABSTRACT

Salinosporamide A ( 1 (NPI-0052)) is a potent, monochlorinated 20S proteasome inhibitor in clinical trials for the treatment of cancer. To elucidate the role of the chlorine leaving group (LG), we synthesized analogues with a range of LG potentials and determined their IC 50 values for inhibition of chymotrypsin-like (CT-L), trypsin-like (T-L), and caspase-like (C-L) activities of 20S proteasomes. Proteasome activity was also determined before and after attempted removal of the inhibitors by dialysis. Analogues bearing substituents with good LG potential exhibited the greatest potency and prolonged duration of proteasome inhibition, with no recovery after 24 h of dialysis. In contrast, activity was restored after

Subject(s)
Lactams/chemical synthesis , Lactams/pharmacology , Lactones/chemical synthesis , Lactones/pharmacology , Proteasome Inhibitors , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Animals , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydrolysis , Kinetics , Lactams/chemistry , Lactones/chemistry , Models, Molecular , Molecular Structure , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Pyrroles/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship
16.
Clin Cancer Res ; 14(16): 5116-23, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18698029

ABSTRACT

PURPOSE: In the current study, we investigate the activation of antiapoptotic signaling pathways in response to proteasome inhibitor treatment in pancreatic cancer and evaluate the use of concomitant inhibition of these pathways to augment proteasome inhibitor treatment responses. EXPERIMENTAL DESIGN: Pancreatic cancer cell lines and mouse flank xenografts were treated with proteasome inhibitor alone or in combination with chemotherapeutic compounds (gemcitabine, erlotinib, and bevacizumab), induction of apoptosis and effects on tumor growth were assessed. The effect of bortezomib (a first-generation proteasome inhibitor) and NPI-0052 (a second-generation proteasome inhibitor) treatment on key pancreatic mitogenic and antiapoptotic pathways [epidermal growth factor receptor, extracellular signal-regulated kinase, and phosphoinositide-3-kinase (PI3K)/AKT] was determined and the ability of inhibitors of these pathways to enhance the effects of proteasome inhibition was assessed in vitro and in vivo. RESULTS: Our data showed that proteasome inhibitor treatment activates antiapoptotic and mitogenic signaling pathways (epidermal growth factor receptor, extracellular signal-regulated kinase, c-Jun-NH2-kinase, and PI3K/AKT) in pancreatic cancer. Additionally, we found that activation of these pathways impairs tumor response to proteasome inhibitor treatment and inhibition of the c-Jun-NH2-kinase and PI3K/AKT pathways increases the antitumor effects of proteasome inhibitor treatment. CONCLUSION: These preclinical studies suggest that targeting proteasome inhibitor-induced antiapoptotic signaling pathways in combination with proteasome inhibition may augment treatment response in highly resistant solid organ malignancies. Further evaluation of these novel treatment combinations in clinical trials is warranted.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , ErbB Receptors/drug effects , Pancreatic Neoplasms/drug therapy , Protease Inhibitors/therapeutic use , Signal Transduction/drug effects , Adenocarcinoma/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Apoptosis/physiology , Bevacizumab , Blotting, Western , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Cetuximab , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Humans , Lactones/administration & dosage , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proteasome Inhibitors , Pyrazines/administration & dosage , Pyrroles/administration & dosage , Quinazolines/administration & dosage , Signal Transduction/physiology , Xenograft Model Antitumor Assays , Gemcitabine
17.
Blood ; 111(9): 4752-63, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18316628

ABSTRACT

Waldenström macroglobulinemia (WM) is an incurable low-grade B-cell lymphoma characterized by high protein turnover. We dissected the biologic role of the proteasome in WM using 2 proteasome inhibitors, NPI-0052 and bortezomib. We found that NPI-0052 inhibited proliferation and induced apoptosis in WM cells, and that the combination of NPI-0052 and bortezomib induced synergistic cytotoxicity in WM cells, leading to inhibition of nuclear translocation of p65NF-kappaB and synergistic induction of caspases-3, -8, and -9 and PARP cleavage. These 2 agents inhibited the canonical and noncanonical NF-kappaB pathways and acted synergistically through their differential effect on Akt activity and on chymotrypsin-like, caspaselike, and trypsinlike activities of the proteasome. We demonstrated that NPI-0052-induced cytotoxicity was completely abrogated in an Akt knockdown cell line, indicating that its major activity is mediated through the Akt pathway. Moreover, we demonstrated that NPI-0052 and bortezomib inhibited migration and adhesion in vitro and homing of WM cells in vivo, and overcame resistance induced by mesenchymal cells or by the addition of interleukin-6 in a coculture in vitro system. Theses studies enhance our understanding of the biologic role of the proteasome pathway in WM, and provide the preclinical basis for clinical trials of combinations of proteasome inhibitors in WM.


Subject(s)
Proteasome Inhibitors , Waldenstrom Macroglobulinemia/drug therapy , Boronic Acids/pharmacology , Bortezomib , Cell Adhesion/drug effects , Cell Death , Cell Movement/drug effects , Cells, Cultured , Drug Delivery Systems , Drug Synergism , Humans , Lactones/pharmacology , Pyrazines/pharmacology , Pyrroles/pharmacology , Waldenstrom Macroglobulinemia/enzymology , Waldenstrom Macroglobulinemia/pathology
18.
Blood ; 111(3): 1654-64, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18006697

ABSTRACT

Our recent study demonstrated that a novel proteasome inhibitor NPI-0052 triggers apoptosis in multiple myeloma (MM) cells, and importantly, that is distinct from bortezomib (Velcade) in its chemical structure, effects on proteasome activities, and mechanisms of action. Here, we demonstrate that combining NPI-0052 and bortezomb induces synergistic anti-MM activity both in vitro using MM cell lines or patient CD138(+) MM cells and in vivo in a human plasmacytoma xenograft mouse model. NPI-0052 plus bortezomib-induced synergistic apoptosis is associated with: (1) activation of caspase-8, caspase-9, caspase-3, and PARP; (2) induction of endoplasmic reticulum (ER) stress response and JNK; (3) inhibition of migration of MM cells and angiogenesis; (4) suppression of chymotrypsin-like (CT-L), caspase-like (C-L), and trypsin-like (T-L) proteolytic activities; and (5) blockade of NF-kappaB signaling. Studies in a xenograft model show that low dose combination of NPI-0052 and bortezomib is well tolerated and triggers synergistic inhibition of tumor growth and CT-L, C-L, and T-L proteasome activities in tumor cells. Immununostaining of MM tumors from NPI-0052 plus bortezomib-treated mice showed growth inhibition, apoptosis, and a decrease in associated angiogenesis. Taken together, our study provides the preclinical rationale for clinical protocols evaluating bortezomib together with NPI-0052 to improve patient outcome in MM.


Subject(s)
Boronic Acids/toxicity , Lactones/toxicity , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Protease Inhibitors/toxicity , Proteasome Endopeptidase Complex/metabolism , Pyrazines/toxicity , Pyrroles/toxicity , Animals , Bortezomib , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Humans , Mice , Multiple Myeloma/blood supply , NF-kappa B/metabolism , Neovascularization, Pathologic , Xenograft Model Antitumor Assays
19.
Blood ; 110(7): 2286-95, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17609425

ABSTRACT

Salinosporamide A (also called NPI-0052), recently identified from the marine bacterium Salinispora tropica, is a potent inhibitor of 20S proteasome and exhibits therapeutic potential against a wide variety of tumors through a poorly understood mechanism. Here we demonstrate that salinosporamide A potentiated the apoptosis induced by tumor necrosis factor alpha (TNF), bortezomib, and thalidomide, and this correlated with down-regulation of gene products that mediate cell proliferation (cyclin D1, cyclooxygenase-2 [COX-2], and c-Myc), cell survival (Bcl-2, Bcl-xL, cFLIP, TRAF1, IAP1, IAP2, and survivin), invasion (matrix metallopro-teinase-9 [MMP-9] and ICAM-1), and angiogenesis (vascular endothelial growth factor [VEGF]). Salinosporamide A also suppressed TNF-induced tumor cell invasion and receptor activator of nuclear factor kappaB ligand (RANKL)-induced osteoclastogenesis. We also found that it suppressed both constitutive and inducible NF-kappaB activation. Compared with bortezomib, MG-132, N-acetyl-leucyl-leucyl-norleucinal (ALLN), and lactacystin, salinosporamide A was found to be the most potent suppressor of NF-kappaB activation. Further studies showed that salinosporamide A inhibited TNF-induced inhibitory subunit of NF-kappaB alpha (IkappaBalpha) degradation, nuclear translocation of p65, and NF-kappaB-dependent reporter gene expression but had no effect on IkappaBalpha kinase activation, IkappaBalpha phosphorylation, or IkappaBalpha ubiquitination. Thus, overall, our results indicate that salinosporamide A enhances apoptosis, suppresses osteoclastogenesis, and inhibits invasion through suppression of the NF-kappaB pathway.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Down-Regulation/drug effects , Lactones/pharmacology , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Pyrroles/pharmacology , Active Transport, Cell Nucleus , Animals , Cell Line , Enzyme Activation , Gene Expression Regulation , Genes, Reporter/genetics , Humans , I-kappa B Proteins/metabolism , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/deficiency , NF-kappa B/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Osteoclasts/cytology , Phosphorylation , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , RANK Ligand/pharmacology , Time Factors , Tumor Necrosis Factors/pharmacology
20.
Mol Pharmacol ; 71(6): 1545-53, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17329499

ABSTRACT

Terpenoids constitute a large family of natural steroids that are widely distributed in plants and insects. We investigated the effects of a series of diterpenes structurally related to acanthoic acid in macrophage functions. We found that diterpenes with different substitutions at the C4 position in ring A are potent activators of liver X receptors (LXRalpha and LXRbeta) in both macrophage cell lines from human and mouse origin and primary murine macrophages. Activation of LXR by these diterpenes was evaluated in transient transfection assays and gene expression analysis of known LXR-target genes, including the cholesterol transporters ABCA1 and ABCG1, the sterol regulatory element-binding protein 1c, and the apoptosis inhibitor of macrophages (Spalpha). Moreover, active diterpenes greatly stimulated cholesterol efflux from macrophages. It is interesting that these diterpenes antagonize inflammatory gene expression mainly through LXR-dependent mechanisms, indicating that these compounds can activate both LXR activation and repression functions. Stimulation of macrophages with acanthoic acid diterpenes induced LXR-target gene expression and cholesterol efflux to similar levels observed with synthetic agonists 3-[3-[N-(2-chloro-3-trifluoromethylbenzyl)-(2,2-diphenylethyl)-amino]propyloxy]phenylacetic acid hydrochloride (GW3965) and N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)-ethyl]phenyl]-benzenesulfonamide [T1317 (T0901317)]. These effects observed in gene expression were deficient in macrophages lacking both LXR isoforms (LXRalpha,beta(-/-)). These results show the ability of certain acanthoic acid diterpenes to activate efficiently both LXRs and suggest that these compounds can exert beneficial effects from a cardiovascular standpoint through LXR-dependent mechanisms.


Subject(s)
DNA-Binding Proteins/metabolism , Diterpenes/pharmacology , Macrophages/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/physiology , Diterpenes/chemistry , Gene Expression , Humans , Liver X Receptors , Macrophages/metabolism , Mice , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/physiology
SELECTION OF CITATIONS
SEARCH DETAIL