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










Publication year range
1.
Sci Transl Med ; 13(611): eaba7791, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34524860

ABSTRACT

SUMOylation, the covalent conjugation of small ubiquitin-like modifier (SUMO) proteins to protein substrates, has been reported to suppress type I interferon (IFN1) responses. TAK-981, a selective small-molecule inhibitor of SUMOylation, pharmacologically reactivates IFN1 signaling and immune responses against cancers. In vivo treatment of wild-type mice with TAK-981 up-regulated IFN1 gene expression in blood cells and splenocytes. Ex vivo treatment of mouse and human dendritic cells promoted their IFN1-dependent activation, and vaccination studies in mice demonstrated stimulation of antigen cross-presentation and T cell priming in vivo. TAK-981 also directly stimulated T cell activation, driving enhanced T cell sensitivity and response to antigen ex vivo. Consistent with these observations, TAK-981 inhibited growth of syngeneic A20 and MC38 tumors in mice, dependent upon IFN1 signaling and CD8+ T cells, and associated with increased intratumoral T and natural killer cell number and activation. Combination of TAK-981 with anti-PD1 or anti-CTLA4 antibodies improved the survival of mice bearing syngeneic CT26 and MC38 tumors. In conclusion, TAK-981 is a first-in-class SUMOylation inhibitor that promotes antitumor immune responses through activation of IFN1 signaling. TAK-981 is currently being studied in phase 1 clinical trials (NCT03648372, NCT04074330, NCT04776018, and NCT04381650) for the treatment of patients with solid tumors and lymphomas.


Subject(s)
Immunity , Sumoylation
2.
J Med Chem ; 64(5): 2501-2520, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33631934

ABSTRACT

SUMOylation is a reversible post-translational modification that regulates protein function through covalent attachment of small ubiquitin-like modifier (SUMO) proteins. The process of SUMOylating proteins involves an enzymatic cascade, the first step of which entails the activation of a SUMO protein through an ATP-dependent process catalyzed by SUMO-activating enzyme (SAE). Here, we describe the identification of TAK-981, a mechanism-based inhibitor of SAE which forms a SUMO-TAK-981 adduct as the inhibitory species within the enzyme catalytic site. Optimization of selectivity against related enzymes as well as enhancement of mean residence time of the adduct were critical to the identification of compounds with potent cellular pathway inhibition and ultimately a prolonged pharmacodynamic effect and efficacy in preclinical tumor models, culminating in the identification of the clinical molecule TAK-981.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Sulfonic Acids/therapeutic use , Sumoylation/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Mice , Molecular Structure , Protein Binding , Protein Processing, Post-Translational/drug effects , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/metabolism , Ubiquitin-Activating Enzymes/metabolism , Xenograft Model Antitumor Assays
3.
Nat Med ; 24(2): 186-193, 2018 02.
Article in English | MEDLINE | ID: mdl-29334375

ABSTRACT

The ubiquitin-proteasome system (UPS) comprises a network of enzymes that is responsible for maintaining cellular protein homeostasis. The therapeutic potential of this pathway has been validated by the clinical successes of a number of UPS modulators, including proteasome inhibitors and immunomodulatory imide drugs (IMiDs). Here we identified TAK-243 (formerly known as MLN7243) as a potent, mechanism-based small-molecule inhibitor of the ubiquitin activating enzyme (UAE), the primary mammalian E1 enzyme that regulates the ubiquitin conjugation cascade. TAK-243 treatment caused depletion of cellular ubiquitin conjugates, resulting in disruption of signaling events, induction of proteotoxic stress, and impairment of cell cycle progression and DNA damage repair pathways. TAK-243 treatment caused death of cancer cells and, in primary human xenograft studies, demonstrated antitumor activity at tolerated doses. Due to its specificity and potency, TAK-243 allows for interrogation of ubiquitin biology and for assessment of UAE inhibition as a new approach for cancer treatment.


Subject(s)
Neoplasms/drug therapy , Nucleosides/pharmacology , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Animals , Cell Line, Tumor , DNA Damage/drug effects , DNA Repair/drug effects , Humans , Imides/pharmacology , Mice , Neoplasms/genetics , Neoplasms/pathology , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Protein Binding , Pyrazoles , Pyrimidines , Sulfides , Ubiquitin/antagonists & inhibitors , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Xenograft Model Antitumor Assays
4.
Bioorg Med Chem Lett ; 26(4): 1156-60, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26804230

ABSTRACT

Investigations of a biaryl ether scaffold identified tetrahydronaphthalene Raf inhibitors with good in vivo activity; however these compounds had affinity toward the hERG potassium channel. Herein we describe our work to eliminate this hERG activity via alteration of the substituents on the benzoic amide functionality. The resulting compounds have improved selectivity against the hERG channel, good pharmacokinetic properties and potently inhibit the Raf pathway in vivo.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Tetrahydronaphthalenes/chemistry , Animals , Cell Line, Tumor , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Inhibitory Concentration 50 , Male , Mice , Mutagenesis , Neoplasms/drug therapy , Neoplasms/pathology , Protein Binding , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tetrahydronaphthalenes/pharmacokinetics , Tetrahydronaphthalenes/therapeutic use , Transplantation, Heterologous
5.
PLoS One ; 10(12): e0144825, 2015.
Article in English | MEDLINE | ID: mdl-26709701

ABSTRACT

In non-clinical studies, the proteasome inhibitor ixazomib inhibits cell growth in a broad panel of solid tumor cell lines in vitro. In contrast, antitumor activity in xenograft tumors is model-dependent, with some solid tumors showing no response to ixazomib. In this study we examined factors responsible for ixazomib sensitivity or resistance using mouse xenograft models. A survey of 14 non-small cell lung cancer (NSCLC) and 6 colon xenografts showed a striking relationship between ixazomib activity and KRAS genotype; tumors with wild-type (WT) KRAS were more sensitive to ixazomib than tumors harboring KRAS activating mutations. To confirm the association between KRAS genotype and ixazomib sensitivity, we used SW48 isogenic colon cancer cell lines. Either KRAS-G13D or KRAS-G12V mutations were introduced into KRAS-WT SW48 cells to generate cells that stably express activated KRAS. SW48 KRAS WT tumors, but neither SW48-KRAS-G13D tumors nor SW48-KRAS-G12V tumors, were sensitive to ixazomib in vivo. Since activated KRAS is known to be associated with metabolic reprogramming, we compared metabolite profiling of SW48-WT and SW48-KRAS-G13D tumors treated with or without ixazomib. Prior to treatment there were significant metabolic differences between SW48 WT and SW48-KRAS-G13D tumors, reflecting higher oxidative stress and glucose utilization in the KRAS-G13D tumors. Ixazomib treatment resulted in significant metabolic regulation, and some of these changes were specific to KRAS WT tumors. Depletion of free amino acid pools and activation of GCN2-eIF2α-pathways were observed both in tumor types. However, changes in lipid beta oxidation were observed in only the KRAS WT tumors. The non-clinical data presented here show a correlation between KRAS genotype and ixazomib sensitivity in NSCLC and colon xenografts and provide new evidence of regulation of key metabolic pathways by proteasome inhibition.


Subject(s)
Boron Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Glycine/analogs & derivatives , Lung Neoplasms/drug therapy , Proteasome Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Amino Acids/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Fatty Acids/metabolism , Glucose Transporter Type 4/biosynthesis , Glycine/therapeutic use , HCT116 Cells , Humans , Lung Neoplasms/metabolism , Metabolome/physiology , Mice , Oxidation-Reduction/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Res ; 73(23): 7043-55, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24121489

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway is particularly important for the survival and proliferation of melanoma cells. Somatic mutations in BRAF and NRAS are frequently observed in melanoma. Recently, the BRAF inhibitors vemurafenib and dabrafenib have emerged as promising agents for the treatment of melanoma patients with BRAF-activating mutations. However, as BRAF inhibitors induce RAF paradoxical activation via RAF dimerization in BRAF wild-type cells, rapid emergence of acquired resistance and secondary skin tumors as well as presence of few effective treatment options for melanoma bearing wild-type BRAF (including NRAS-mutant melanoma) are clinical concerns. Here, we demonstrate that the selective pan-RAF inhibitor TAK-632 suppresses RAF activity in BRAF wild-type cells with minimal RAF paradoxical activation. Our analysis using RNAi and TAK-632 in preclinical models reveals that the MAPK pathway of NRAS-mutated melanoma cells is highly dependent on RAF. We also show that TAK-632 induces RAF dimerization but inhibits the kinase activity of the RAF dimer, probably because of its slow dissociation from RAF. As a result, TAK-632 demonstrates potent antiproliferative effects both on NRAS-mutated melanoma cells and BRAF-mutated melanoma cells with acquired resistance to BRAF inhibitors through NRAS mutation or BRAF truncation. Furthermore, we demonstrate that the combination of TAK-632 and the MAPK kinase (MEK) inhibitor TAK-733 exhibits synergistic antiproliferative effects on these cells. Our findings characterize the unique features of TAK-632 as a pan-RAF inhibitor and provide rationale for its further investigation in NRAS-mutated melanoma and a subset of BRAF-mutated melanomas refractory to BRAF inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzothiazoles/therapeutic use , Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Nitriles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , raf Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Cells, Cultured , Humans , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Mice , Mice, Nude , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Cancer Res ; 71(15): 5336-45, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21646478

ABSTRACT

Activation of the Sonic hedgehog (Shh) pathway and increased expression of Gli1 play an important role in proliferation and transformation of granule cell progenitors (GCP) in the developing cerebellum. Medulloblastomas arising from cerebellar GCPs are frequently driven by Shh pathway-activating mutations; however, molecular mechanisms of Shh pathway dysregulation and transformation of neural progenitors remain poorly defined. We report that the transcription factor and oncogene Snail1 (Sna1) is directly induced by Shh pathway activity in GCPs, murine medulloblastomas, and human medulloblastoma cells. Enforced expression of Sna1 was sufficient to induce GCPs and medulloblastoma cell proliferation in the absence of Shh/Gli1 exposure. In addition, enforced expression of Sna1 increased transformation of medulloblastoma cells in vitro and in vivo. Analysis of potential Sna1 targets in neural cells revealed a novel Sna1 target, N-Myc, a transcription factor known to play a role in Shh-mediated GCP proliferation and medulloblastoma formation. We found that Sna1 directly induced transcription of N-Myc in human medulloblastoma cells and that depletion of N-Myc ablated the Sna1-induced proliferation and transformation. Taken together, these results provide further insight into the mechanism of Shh-induced transformation of neural progenitor cells and suggest that induction of Sna1 may serve to amplify the oncogenic potential of Shh pathway activation through N-Myc induction.


Subject(s)
Cell Transformation, Neoplastic , Cerebellar Neoplasms/pathology , Hedgehog Proteins/physiology , Medulloblastoma/pathology , Neurons/cytology , Proto-Oncogene Proteins c-myc/physiology , Transcription Factors/physiology , Animals , Cell Division , Cell Line, Tumor , Cerebellum/cytology , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Mice , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Zinc Finger Protein GLI1
8.
J Med Chem ; 54(6): 1836-46, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21341678

ABSTRACT

Inhibition of mutant B-Raf signaling, through either direct inhibition of the enzyme or inhibition of MEK, the direct substrate of Raf, has been demonstrated preclinically to inhibit tumor growth. Very recently, treatment of B-Raf mutant melanoma patients with a selective B-Raf inhibitor has resulted in promising preliminary evidence of antitumor activity. This article describes the design and optimization of tetrahydronaphthalene-derived compounds as potent inhibitors of the Raf pathway in vitro and in vivo. These compounds possess good pharmacokinetic properties in rodents and inhibit B-Raf mutant tumor growth in mouse xenograft models.


Subject(s)
Antineoplastic Agents/chemical synthesis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Tetrahydronaphthalenes/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Availability , Crystallography, X-Ray , Drug Design , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Mice, Nude , Models, Molecular , Mutation , Proto-Oncogene Proteins B-raf/genetics , Stereoisomerism , Structure-Activity Relationship , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/pharmacology , Xenograft Model Antitumor Assays
9.
Cancer Res ; 71(3): 675-85, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21148750

ABSTRACT

The mitotic kinase Aurora A is an important therapeutic target for cancer therapy. This study evaluated new mechanism-based pharmacodynamic biomarkers in cancer patients in two phase I studies of MLN8054, a small-molecule inhibitor of Aurora A kinase. Patients with advanced solid tumors received MLN8054 orally for 7 consecutive days in escalating dose cohorts, with skin and tumor biopsies obtained before and after dosing. Skin biopsies were evaluated for increased mitotic cells within the basal epithelium. Tumor biopsies were assessed for accumulation of mitotic cells within proliferative tumor regions. Several patients in the highest dose cohorts showed marked increases in the skin mitotic index after dosing. Although some tumors exhibited increases in mitotic cells after dosing, others displayed decreases, a variable outcome consistent with dual mechanisms of mitotic arrest and mitotic slippage induced by antimitotics in tumors. To provide a clearer picture, mitotic cell chromosome alignment and spindle bipolarity, new biomarkers of Aurora A inhibition that act independently of mitotic arrest or slippage, were assessed in the tumor biopsies. Several patients, primarily in the highest dose cohorts, had marked decreases in the percentage of mitotic cells with aligned chromosomes and bipolar spindles after dosing. Evidence existed for an exposure-effect relationship for mitotic cells with defects in chromosome alignment and spindle bipolarity that indicated a biologically active dose range. Outcomes of pharmacodynamic assays from skin and tumor biopsies were concordant in several patients. Together, these new pharmacodynamic assays provide evidence for Aurora A inhibition by MLN8054 in patient skin and tumor tissues.


Subject(s)
Benzazepines/pharmacology , Benzazepines/pharmacokinetics , Biomarkers, Tumor/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Aurora Kinases , Benzazepines/adverse effects , Benzazepines/blood , Biomarkers, Tumor/blood , Biopsy , Dose-Response Relationship, Drug , Humans , Mitosis/drug effects , Neoplasms/blood , Neoplasms/pathology , Skin/metabolism , Skin/pathology
10.
Cancer Chemother Pharmacol ; 67(4): 945-54, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20607239

ABSTRACT

PURPOSE: Aurora A kinase is critical in assembly and function of the mitotic spindle. It is overexpressed in various tumor types and implicated in oncogenesis and tumor progression. This trial evaluated the dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) of MLN8054, a selective small-molecule inhibitor of Aurora A kinase. METHODS: In this first-in-human, dose-escalation study, MLN8054 was given orally for 7, 14, or 21 days followed by a 14-day treatment-free period. Escalating cohorts of 3-6 patients with advanced solid tumors were treated until DLT was seen in ≥2 patients in a cohort. Serial blood samples were collected for pharmacokinetics and skin biopsies were collected for pharmacodynamics. RESULTS: Sixty-one patients received 5, 10, 20, 30, or 40 mg once daily for 7 days; 25, 35, 45, or 55 mg/day in four divided doses (QID) for 7 days; or 55, 60, 70, or 80 mg/day plus methylphenidate or modafinil with daytime doses (QID/M) for 7-21 days. DLTs of reversible grade 3 benzodiazepine-like effects defined the estimated MTD of 60 mg QID/M for 14 days. MLN8054 was absorbed rapidly, exposure was dose proportional, and terminal half-life was 30-40 h. Three patients had stable disease for >6 cycles. CONCLUSIONS: MLN8054 dosing for up to 14 days of a 28-day cycle was feasible. Reversible somnolence was dose limiting and prevented achievement of plasma concentrations predicted necessary for target modulation. A recommended dose for investigation in phase 2 trials was not established. A second-generation Aurora A kinase inhibitor is in development.


Subject(s)
Antineoplastic Agents/adverse effects , Benzazepines/adverse effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Aurora Kinases , Benzazepines/administration & dosage , Benzazepines/pharmacokinetics , Benzhydryl Compounds/therapeutic use , Central Nervous System Stimulants/therapeutic use , Disorders of Excessive Somnolence/chemically induced , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Methylphenidate/therapeutic use , Middle Aged , Modafinil , Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Young Adult
11.
Bioorg Med Chem Lett ; 20(16): 4800-4, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20634068

ABSTRACT

The discovery of novel pyrazoline derivatives as B-Raf (V600E) inhibitors is described in this report. Chemical modification of the pyrazoline scaffold led to the development of SAR and identified potent and selective inhibitors of B-Raf (V600E). Determination of the pharmacokinetic properties of selected inhibitors is also reported.


Subject(s)
Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyrazoles/chemistry , Amino Acid Substitution , Binding Sites , Computer Simulation , Drug Evaluation, Preclinical , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
12.
J Biol Chem ; 285(26): 19747-56, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20427282

ABSTRACT

Members of the transforming growth factor-beta superfamily play essential roles in both the pluripotency and differentiation of embryonic stem (ES) cells. Although bone morphogenic proteins (BMPs) maintain pluripotency of undifferentiated mouse ES cells, the role of autocrine Nodal signaling is less clear. Pharmacological, molecular, and genetic methods were used to further understand the roles and potential interactions of these pathways. Treatment of undifferentiated ES cells with SB431542, a pharmacological inhibitor of Smad2 signaling, resulted in a rapid reduction of phosphorylated Smad2 and altered the expression of several putative downstream targets. Unexpectedly, inhibition of the Nodal signaling pathway resulted in enhanced BMP signaling, as assessed by Smad1/5 phosphorylation. SB431542-treated cells also demonstrated significant induction of the Id genes, which are known direct targets of BMP signaling and important factors in ES cell pluripotency. Inhibition of BMP signaling decreased the SB431542-mediated phosphorylation of Smad1/5 and induction of Id genes, suggesting that BMP signaling is necessary for some Smad2-mediated activity. Because Smad7, a known inhibitory factor to both Nodal and BMP signaling, was down-regulated following inhibition of Nodal-Smad2 signaling, the contribution of Smad7 to the cross-talk between the transforming growth factor-beta pathways in ES cells was examined. Biochemical manipulation of Smad7 expression, through shRNA knockdown or inducible gene expression, significantly reduced the SB431542-mediated phosphorylation of Smad1/5 and induction of the Id genes. We conclude that autocrine Nodal signaling in undifferentiated mouse ES cells modulates the vital pluripotency pathway of BMP signaling.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Embryonic Stem Cells/metabolism , Nodal Protein/metabolism , Signal Transduction , Animals , Autocrine Communication , Benzamides/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Dioxoles/pharmacology , Embryonic Stem Cells/cytology , Female , Gene Expression/drug effects , Immunoblotting , Left-Right Determination Factors/genetics , Left-Right Determination Factors/metabolism , Male , Mice , Mice, Knockout , Nodal Protein/genetics , Phosphorylation/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism
13.
Tissue Eng Part A ; 15(7): 1797-805, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19191513

ABSTRACT

Biodegradable polymer scaffolds provide an excellent approach to quantifying critical factors necessary for restoration of function after a transection spinal cord injury. Neural stem cells (NSCs) and Schwann cells (SCs) support axonal regeneration. This study examines the compatibility of NSCs and SCs with the poly-lactic-co-glycolic acid polymer scaffold and quantitatively assesses their potential to promote regeneration after a spinal cord transection injury in rats. NSCs were cultured as neurospheres and characterized by immunostaining for nestin (NSCs), glial fibrillary acidic protein (GFAP) (astrocytes), betaIII-tubulin (immature neurons), oligodendrocyte-4 (immature oligodendrocytes), and myelin oligodendrocyte (mature oligodendrocytes), while SCs were characterized by immunostaining for S-100. Rats with transection injuries received scaffold implants containing NSCs (n=17), SCs (n=17), and no cells (control) (n=8). The degree of axonal regeneration was determined by counting neurofilament-stained axons through the scaffold channels 1 month after transplantation. Serial sectioning through the scaffold channels in NSC- and SC-treated groups revealed the presence of nestin, neurofilament, S-100, and betaIII tubulin-positive cells. GFAP-positive cells were only seen at the spinal cord-scaffold border. There were significantly more axons in the NSC- and SC- treated groups compared to the control group. In conclusion, biodegradable scaffolds with aligned columns seeded with NSCs or SCs facilitate regeneration across the transected spinal cord. Further, these multichannel biodegradable polymer scaffolds effectively serve as platforms for quantitative analysis of axonal regeneration.


Subject(s)
Axons/physiology , Biocompatible Materials/metabolism , Lactic Acid/metabolism , Neurons/cytology , Polyglycolic Acid/metabolism , Schwann Cells/cytology , Spinal Cord Injuries/physiopathology , Stem Cells/cytology , Animals , Animals, Newborn , Axons/metabolism , Axons/pathology , Cell Shape , Cell Survival , Cells, Cultured , Nerve Regeneration , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Recovery of Function , Schwann Cells/metabolism , Schwann Cells/transplantation , Stem Cell Transplantation , Survival Analysis , Tissue Scaffolds
14.
Stem Cells ; 26(4): 1027-36, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18276799

ABSTRACT

Sonic hedgehog (Shh) is necessary for sustaining the proliferation of neural stem cells (NSCs), yet little is known about its mechanisms. Whereas Gli1, Gli2, and Gli3, the primary mediators of Shh signaling, were all expressed in hippocampal neural progenitors, Shh treatment of NSCs induced only Gli1 expression. Acute depletion of Gli1 in postnatal NSCs by short-hairpin RNA decreased proliferation, whereas germline deletion of Gli1 did not affect NSC proliferation, suggesting a difference in mechanisms of Gli1 compensation that may be developmentally dependent. To determine whether Gli1 was sufficient to enhance NSC proliferation, we overexpressed this mitogen and were surprised to find that Gli1 resulted in decreased proliferation, accumulation of NSCs in the G2/M phase of cell cycle, and apoptosis. In contrast, Gli1-expressing lineage-restricted neural precursors demonstrated a 4.5-fold proliferation enhancement. Expression analyses of Gli1-expressing NSCs identified significant induction of Gadd45a and decreased cyclin A2 and Stag1 mRNA, genes involved in the G2-M transition and apoptosis. Furthermore, Gadd45a overexpression was sufficient to partially recapitulate the Gli1-induced G2/M accumulation and cell death of NSCs. In contrast to normal stem cells, tumor-derived stem cells had markedly higher basal Gli1 expression and did not undergo apoptosis with further elevation of Gli1. Our data suggest that Gli1-induced apoptosis may serve as a protective mechanism against premature mitosis and may give insight into mechanisms by which nonmalignant stem cells restrain hyperproliferation in the context of potentially transforming mitogenic signals. Tumor-derived stem cells apparently lack these mechanisms, which may contribute to their unrestrained proliferation and malignant potential.


Subject(s)
Apoptosis/physiology , Cell Division/physiology , G2 Phase/physiology , Hippocampus/physiology , Kruppel-Like Transcription Factors/physiology , Neoplastic Stem Cells/pathology , Neurons/pathology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Division/genetics , Cells, Cultured , G2 Phase/genetics , Hippocampus/metabolism , Hippocampus/pathology , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/deficiency , Intermediate Filament Proteins/genetics , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nestin , Neurons/cytology , Neurons/metabolism , Tumor Cells, Cultured , Zinc Finger Protein GLI1
15.
Cancer Res ; 67(11): 5362-70, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545617

ABSTRACT

Aurora A is a serine/threonine protein kinase essential for normal mitotic progression. Aberrant increased expression of Aurora A, which occurs frequently in human cancers, results in abnormal mitoses leading to chromosome instability and possibly tumorigenesis. Consequently, Aurora A has received considerable attention as a potential target for anticancer therapeutic intervention. Aurora A coordinates several essential mitotic activities through phosphorylation of a variety of proteins, including TACC3, which modulates microtubule stabilization of the mitotic spindle. Recent studies identified a conserved serine in Xenopus (Ser(626)) and Drosophila (Ser(863)) TACC3 orthologues that is phosphorylated by Aurora A. We show that this conserved serine on human TACC3 (Ser(558)) is also phosphorylated by Aurora A. Moreover, phosphorylation of TACC3 by Aurora A in human cells is essential for its proper localization to centrosomes and proximal mitotic spindles. Inhibition of Aurora A with the selective small molecule inhibitor MLN8054 in cultured human tumor cells resulted in mislocalization of TACC3 away from mitotic spindles in a concentration-dependent manner. Furthermore, oral administration of MLN8054 to nude mice bearing HCT-116 human tumor xenografts caused a dose-dependent mislocalization of TACC3 away from spindle poles that correlated with tumor growth inhibition. As TACC3 localization to mitotic spindles depends on Aurora A-mediated phosphorylation, quantifying TACC3 mislocalization represents a novel pharmacodynamic approach for measuring Aurora A activity in cancer patients treated with inhibitors of Aurora A kinase.


Subject(s)
Benzazepines/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence , Animals , Aurora Kinase A , Aurora Kinases , Centrosome/metabolism , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Serine/metabolism , Xenograft Model Antitumor Assays
16.
Dev Biol ; 308(2): 331-42, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17599824

ABSTRACT

Sonic hedgehog (Shh), a secreted morphogen and mitogen, is essential for nervous system development and neural stem cell (NSC) self-renewal. As the intracellular signal transduction of Shh in NSCs is largely unknown, we sought to characterize pathway targets using ligand stimulation and genetic models of activation. NSCs haploinsufficient for Patched (Ptc), a receptor repressive to Shh signaling, showed enhanced proliferation of a magnitude similar to Shh-treated wild-type (Wt) NSCs. Analysis of the Gli zinc-finger transcription factors, primary mediators of Shh activity, demonstrated differential induction between models of pathway activation. Gli1 was significantly induced in Wt NSCs exposed to Shh, whereas Gli2 was elevated and Gli1 expression did not change in Ptc(+/-) NSCs. Other Shh targets (Nmyc, Id factors) were induced under both conditions of pathway activation. Interestingly, Shh-treated Ptc(+/-) NSCs induced expression of Gli1 but failed to increase proliferation, suggesting that the NSCs may have reached a physiologic plateau in proliferative capacity. Thus, our data demonstrate that Ptc(+/-) mice have an expanded progenitor cell niche in vivo and that NSCs maintain a cell-intrinsic increase in basal proliferation in vitro that is sustained by a Gli transduction signature distinct from that of exogenous Shh stimulation. Additionally, Ptc(+/-) NSCs maintain tight control over mitosis and do not further augment proliferation in the presence of mitogenic stimulation.


Subject(s)
Hedgehog Proteins/metabolism , Multipotent Stem Cells/metabolism , Neurons/metabolism , Animals , Base Sequence , Cell Proliferation/drug effects , Cells, Cultured , DNA Primers/genetics , Gene Expression Regulation, Developmental/drug effects , Genes, myc , Hedgehog Proteins/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Inhibitor of Differentiation Proteins/genetics , Ligands , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Models, Genetic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Transcriptional Activation
17.
Proc Natl Acad Sci U S A ; 104(10): 4106-11, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17360485

ABSTRACT

Increased Aurora A expression occurs in a variety of human cancers and induces chromosomal abnormalities during mitosis associated with tumor initiation and progression. MLN8054 is a selective small-molecule Aurora A kinase inhibitor that has entered Phase I clinical trials for advanced solid tumors. MLN8054 inhibits recombinant Aurora A kinase activity in vitro and is selective for Aurora A over the family member Aurora B in cultured cells. MLN8054 treatment results in G(2)/M accumulation and spindle defects and inhibits proliferation in multiple cultured human tumor cells lines. Growth of human tumor xenografts in nude mice was dramatically inhibited after oral administration of MLN8054 at well tolerated doses. Moreover, the tumor growth inhibition was sustained after discontinuing MLN8054 treatment. In human tumor xenografts, MLN8054 induced mitotic accumulation and apoptosis, phenotypes consistent with inhibition of Aurora A. MLN8054 is a selective inhibitor of Aurora A kinase that robustly inhibits growth of human tumor xenografts and represents an attractive modality for therapeutic intervention of human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Benzazepines/pharmacology , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Disease Progression , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Nude , Neoplasm Transplantation
18.
Behav Neurosci ; 118(1): 163-72, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14979793

ABSTRACT

The "waist" area and external subnuclei of the parabrachial nucleus (PBN) have been implicated in the processing of gustatory information, yet their behavioral roles are not clearly defined. In the current study, areas within and surrounding the PBN were stimulated while oromotor behaviors were monitored in conscious rats. Electrical and chemical (100 mM glutamate) stimulation of the waist area increased ingestive oromotor behaviors over baseline (p<.01). Stimulation of external PBN subnuclei and areas medial and ventral to the PBN failed to cause a behavioral change. These data support the hypothesis that the waist area of the PBN constitutes part of the neural substrate involved in eliciting oromotor behaviors in response to taste input.


Subject(s)
Motor Activity/physiology , Mouth/physiology , Pons/physiology , Animals , Electric Stimulation/methods , Glutamic Acid/pharmacology , Male , Motor Activity/drug effects , Mouth/drug effects , Pons/drug effects , Rats , Rats, Wistar
19.
J Comp Neurol ; 465(2): 296-308, 2003 Oct 13.
Article in English | MEDLINE | ID: mdl-12949788

ABSTRACT

The distribution of quinine-stimulated Fos-like immunoreactivity (FLI) in several subdivisions of the parabrachial nucleus (PBN) known to be responsive to gustatory stimulation was examined in rats in which the chorda tympani nerve (CT) and/or glossopharyngeal nerve (GL) was transected (Experiment 1) and in rats in which the GL was transected with regeneration promoted or prevented (Experiment 2). We confirmed previous findings in the literature by demonstrating that rats intraorally infused with 3 mM quinine showed a robust population of FLI in the waist area and the external lateral (EL) and external medial (EM) subdivisions of the PBN (Yamamoto et al. [1994] Physiol Behav 56:1197-1202; Travers et al., [ 1999] Am J Physiol 277:R384-R394). In the waist area, only GL transection significantly decreased the number of FLI-neurons elicited by intraoral infusion of quinine compared with water-stimulated controls. In the external subdivisions neither neurotomy affected the number of FLI-neurons. The effect of GL transection in the waist area was enduring for rats in which the GL did not regenerate (up to 94 days), but regeneration of the GL after 52 days restored quinine-stimulated FLI to control values. In these same GL-transected animals, there were parallel decreases in the number of gapes elicited by intraoral quinine stimulation that recovered, but only subsequent to regeneration of the GL. These data provide support for the role of the waist area in the brainstem processing that underlies oromotor rejection behaviors and also help substantiate the hypothesis that the CT and GL are relatively specialized with regard to function. Moreover, when the quinine-induced pattern of neural activity in the second central gustatory relay, as assessed by FLI, is substantially altered by the loss of peripheral gustatory input from the GL, it can be restored upon regeneration of the nerve.


Subject(s)
Chorda Tympani Nerve/physiology , Glossopharyngeal Nerve/physiology , Nerve Regeneration/physiology , Oncogene Proteins v-fos/metabolism , Pons/physiology , Taste/physiology , Animals , Axotomy , Immunohistochemistry , Male , Muscle Relaxants, Central/pharmacology , Oncogene Proteins v-fos/drug effects , Quinine/pharmacology , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Tongue/innervation , Tongue/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...