Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Blood Cancer Discov ; 2(3): 266-287, 2021 05.
Article in English | MEDLINE | ID: mdl-34027418

ABSTRACT

We discovered that the survival and growth of many primary acute myeloid leukemia (AML) samples and cell lines, but not normal CD34+ cells, are dependent on SIRT5, a lysine deacylase implicated in regulating multiple metabolic pathways. Dependence on SIRT5 is genotype-agnostic and extends to RAS- and p53-mutated AML. Results were comparable between SIRT5 knockdown and SIRT5 inhibition using NRD167, a potent and selective SIRT5 inhibitor. Apoptosis induced by SIRT5 disruption is preceded by reductions in oxidative phosphorylation and glutamine utilization, and an increase in mitochondrial superoxide that is attenuated by ectopic superoxide dismutase 2. These data indicate that SIRT5 controls and coordinates several key metabolic pathways in AML and implicate SIRT5 as a vulnerability in AML.


Subject(s)
Leukemia, Myeloid, Acute , Sirtuins , Apoptosis , Humans , Leukemia, Myeloid, Acute/drug therapy , Lysine/metabolism , Mitochondria/genetics , Oxidative Phosphorylation , Sirtuins/genetics
2.
Acta Haematol ; 144(4): 458-464, 2021.
Article in English | MEDLINE | ID: mdl-33412552

ABSTRACT

Normal human bone marrow cells are critical for studies of hematopoiesis and as controls to assess toxicity. As cells from commercial vendors are expensive, many laboratories resort to cancer-free bone marrow specimens obtained during staging or to umbilical cord blood cells, which may be abnormal or reflect a much younger age group compared to the disease samples under study. We piloted the use of femoral heads as an alternative and inexpensive source of normal bone marrow. Femoral heads were obtained from 21 successive patients undergoing elective hip arthroplasty. Mononuclear cells (MNCs) were purified with Ficoll, and CD3+, CD14+, and CD34+ cells were purified with antibody-coated microbeads. The median yield of MNCs was 8.95 × 107 (range, 1.62 × 105-2.52 × 108), and the median yield of CD34+ cells was 1.40 × 106 (range, 3.60 × 105-9.90 × 106). Results of downstream applications including qRT-PCR, colony-forming assays, and ex vivo proliferation analysis were of high quality and comparable to those obtained with standard bone marrow aspirates. We conclude that femoral heads currently discarded as medical waste are a cost-efficient source of bone marrow cells for research use.


Subject(s)
Femur Head/cytology , Hematopoietic Stem Cells/cytology , Adult , Aged , Aged, 80 and over , Antigens, CD34/metabolism , Arthroplasty, Replacement, Hip , Case-Control Studies , Fetal Blood/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Middle Aged
4.
Nat Commun ; 10(1): 1784, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992455

ABSTRACT

The incomplete identification of structural variants (SVs) from whole-genome sequencing data limits studies of human genetic diversity and disease association. Here, we apply a suite of long-read, short-read, strand-specific sequencing technologies, optical mapping, and variant discovery algorithms to comprehensively analyze three trios to define the full spectrum of human genetic variation in a haplotype-resolved manner. We identify 818,054 indel variants (<50 bp) and 27,622 SVs (≥50 bp) per genome. We also discover 156 inversions per genome and 58 of the inversions intersect with the critical regions of recurrent microdeletion and microduplication syndromes. Taken together, our SV callsets represent a three to sevenfold increase in SV detection compared to most standard high-throughput sequencing studies, including those from the 1000 Genomes Project. The methods and the dataset presented serve as a gold standard for the scientific community allowing us to make recommendations for maximizing structural variation sensitivity for future genome sequencing studies.


Subject(s)
Genome, Human/genetics , Genomic Structural Variation , Genomics/methods , Haplotypes/genetics , Algorithms , Chromosome Mapping/methods , Databases, Genetic , High-Throughput Nucleotide Sequencing/methods , Humans , INDEL Mutation , Whole Genome Sequencing/methods
5.
J Med Chem ; 62(5): 2651-2665, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30776234

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, incurable cancer with a 20% 1 year survival rate. While standard-of-care therapy can prolong life in a small fraction of cases, PDAC is inherently resistant to current treatments, and novel therapies are urgently required. Histone deacetylase (HDAC) inhibitors are effective in killing pancreatic cancer cells in in vitro PDAC studies, and although there are a few clinical studies investigating combination therapy including HDAC inhibitors, no HDAC drug or combination therapy with an HDAC drug has been approved for the treatment of PDAC. We developed an inhibitor of HDACs, AES-135, that exhibits nanomolar inhibitory activity against HDAC3, HDAC6, and HDAC11 in biochemical assays. In a three-dimensional coculture model, AES-135 kills low-passage patient-derived tumor spheroids selectively over surrounding cancer-associated fibroblasts and has excellent pharmacokinetic properties in vivo. In an orthotopic murine model of pancreatic cancer, AES-135 prolongs survival significantly, therefore representing a candidate for further preclinical testing.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Hydroxamic Acids/chemistry , Pancreatic Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Benzamides/chemistry , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Disease Models, Animal , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/pharmacokinetics , Hydrocarbons, Fluorinated/therapeutic use , Mice , Pancreatic Neoplasms/pathology , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
6.
Clin Cancer Res ; 25(7): 2323-2335, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30563936

ABSTRACT

PURPOSE: Myelofibrosis is a hematopoietic stem cell neoplasm characterized by bone marrow reticulin fibrosis, extramedullary hematopoiesis, and frequent transformation to acute myeloid leukemia. Constitutive activation of JAK/STAT signaling through mutations in JAK2, CALR, or MPL is central to myelofibrosis pathogenesis. JAK inhibitors such as ruxolitinib reduce symptoms and improve quality of life, but are not curative and do not prevent leukemic transformation, defining a need to identify better therapeutic targets in myelofibrosis. EXPERIMENTAL DESIGN: A short hairpin RNA library screening was performed on JAK2V617F-mutant HEL cells. Nuclear-cytoplasmic transport (NCT) genes including RAN and RANBP2 were among top candidates. JAK2V617F-mutant cell lines, human primary myelofibrosis CD34+ cells, and a retroviral JAK2V617F-driven myeloproliferative neoplasms mouse model were used to determine the effects of inhibiting NCT with selective inhibitors of nuclear export compounds KPT-330 (selinexor) or KPT-8602 (eltanexor). RESULTS: JAK2V617F-mutant HEL, SET-2, and HEL cells resistant to JAK inhibition are exquisitely sensitive to RAN knockdown or pharmacologic inhibition by KPT-330 or KPT-8602. Inhibition of NCT selectively decreased viable cells and colony formation by myelofibrosis compared with cord blood CD34+ cells and enhanced ruxolitinib-mediated growth inhibition and apoptosis, both in newly diagnosed and ruxolitinib-exposed myelofibrosis cells. Inhibition of NCT in myelofibrosis CD34+ cells led to nuclear accumulation of p53. KPT-330 in combination with ruxolitinib-normalized white blood cells, hematocrit, spleen size, and architecture, and selectively reduced JAK2V617F-mutant cells in vivo. CONCLUSIONS: Our data implicate NCT as a potential therapeutic target in myelofibrosis and provide a rationale for clinical evaluation in ruxolitinib-exposed patients with myelofibrosis.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Primary Myelofibrosis/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biological Transport/drug effects , Biomarkers , Cell Line, Tumor , Cell Nucleus/drug effects , Computational Biology/methods , Cytoplasm/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Molecular Targeted Therapy , Mutation , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/etiology , STAT Transcription Factors/metabolism , Transcriptome
7.
Leukemia ; 32(11): 2399-2411, 2018 11.
Article in English | MEDLINE | ID: mdl-29749399

ABSTRACT

Tumor necrosis factor alpha (TNF) is increased in myelofibrosis (MF) and promotes survival of malignant over normal cells. The mechanisms altering TNF responsiveness in MF cells are unknown. We show that the proportion of marrow (BM) cells expressing TNF is increased in MF compared to controls, with the largest differential in primitive cells. Blockade of TNF receptor 2 (TNFR2), but not TNFR1, selectively inhibited colony formation by MF CD34+ and mouse JAK2V617F progenitor cells. Microarray of mouse MPN revealed reduced expression of X-linked inhibitor of apoptosis (Xiap) and mitogen-activated protein kinase 8 (Mapk8) in JAK2V617F relative to JAK2WT cells, which were normalized by TNFR2 but not TNFR1 blockade. XIAP and MAPK8 were also reduced in MF CD34+ cells compared to normal BM, and their ectopic expression induced apoptosis. Unlike XIAP, expression of cellular IAP (cIAP) protein was increased in MF CD34+ cells. Consistent with cIAP's role in NF-κB activation, TNF-induced NF-κB activity was higher in MF vs. normal BM CD34+ cells. This suggests that JAK2V617F reprograms TNF response toward survival by downregulating XIAP and MAPK8 through TNFR2. Our results reveal an unexpected pro-apoptotic role for XIAP in MF and identify TNFR2 as a key mediator of TNF-induced clonal expansion.


Subject(s)
Autocrine Communication/physiology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antigens, CD/metabolism , Apoptosis/physiology , Humans , Janus Kinase 2/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism
8.
ChemMedChem ; 11(8): 850-61, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27028877

ABSTRACT

Pharmacologic blockade of the activation of signal transducer and activator of transcription 3 (STAT3) in tyrosine kinase inhibitor (TKI)-resistant chronic myeloid leukemia (CML) cell lines characterized by kinase-independent resistance was shown to re-sensitize CML cells to TKI therapy, suggesting that STAT3 inhibitors in combination with TKIs are an effective combinatorial therapeutic for the treatment of CML. Benzoic acid- and hydroxamic acid-based STAT3 inhibitors SH-4-054 and SH-5-007, developed previously in our laboratory, demonstrated promising activity against these resistant CML cell lines. However, pharmacokinetic studies in murine models (CD-1 mice) revealed that both SH-4-054 and SH-5-007 are susceptible to glutathione conjugation at the para position of the pentafluorophenyl group via nucleophilic aromatic substitution (SN Ar). To determine whether the electrophilicity of the pentafluorophenyl sulfonamide could be tempered, an in-depth structure-activity relationship (SAR) study of the SH-4-054 scaffold was conducted. These studies revealed that AM-1-124, possessing a 2,3,5,6-tetrafluorophenylsulfonamide group, retained STAT3 protein affinity (Ki =15 µm), as well as selectivity over STAT1 (Ki >250 µm). Moreover, in both hepatocytes and in in vivo pharmacokinetic studies (CD-1 mice), AM-1-124 was found to be dramatically more stable than SH-4-054 (t1/2 =1.42 h cf. 10 min, respectively). AM-1-124 is a promising STAT3-targeting inhibitor with demonstrated bioavailability, suitable for evaluation in preclinical cancer models.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Drug Resistance, Neoplasm/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Sulfonamides/pharmacology , para-Aminobenzoates/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , para-Aminobenzoates/chemical synthesis , para-Aminobenzoates/chemistry
9.
Nat Biotechnol ; 34(3): 303-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829319

ABSTRACT

Haplotyping of human chromosomes is a prerequisite for cataloguing the full repertoire of genetic variation. We present a microfluidics-based, linked-read sequencing technology that can phase and haplotype germline and cancer genomes using nanograms of input DNA. This high-throughput platform prepares barcoded libraries for short-read sequencing and computationally reconstructs long-range haplotype and structural variant information. We generate haplotype blocks in a nuclear trio that are concordant with expected inheritance patterns and phase a set of structural variants. We also resolve the structure of the EML4-ALK gene fusion in the NCI-H2228 cancer cell line using phased exome sequencing. Finally, we assign genetic aberrations to specific megabase-scale haplotypes generated from whole-genome sequencing of a primary colorectal adenocarcinoma. This approach resolves haplotype information using up to 100 times less genomic DNA than some methods and enables the accurate detection of structural variants.


Subject(s)
Haplotypes/genetics , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Sequence Analysis, DNA/methods , DNA/genetics , Genome, Human , Genomic Structural Variation , Germ Cells , Humans , Nucleic Acid Conformation , Oncogene Proteins, Fusion/genetics , Polymorphism, Single Nucleotide
10.
Blood ; 125(11): 1772-81, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25573989

ABSTRACT

The mechanisms underlying tyrosine kinase inhibitor (TKI) resistance in chronic myeloid leukemia (CML) patients lacking explanatory BCR-ABL1 kinase domain mutations are incompletely understood. To identify mechanisms of TKI resistance that are independent of BCR-ABL1 kinase activity, we introduced a lentiviral short hairpin RNA (shRNA) library targeting ∼5000 cell signaling genes into K562(R), a CML cell line with BCR-ABL1 kinase-independent TKI resistance expressing exclusively native BCR-ABL1. A customized algorithm identified genes whose shRNA-mediated knockdown markedly impaired growth of K562(R) cells compared with TKI-sensitive controls. Among the top candidates were 2 components of the nucleocytoplasmic transport complex, RAN and XPO1 (CRM1). shRNA-mediated RAN inhibition or treatment of cells with the XPO1 inhibitor, KPT-330 (Selinexor), increased the imatinib sensitivity of CML cell lines with kinase-independent TKI resistance. Inhibition of either RAN or XPO1 impaired colony formation of CD34(+) cells from newly diagnosed and TKI-resistant CML patients in the presence of imatinib, without effects on CD34(+) cells from normal cord blood or from a patient harboring the BCR-ABL1(T315I) mutant. These data implicate RAN in BCR-ABL1 kinase-independent imatinib resistance and show that shRNA library screens are useful to identify alternative pathways critical to drug resistance in CML.


Subject(s)
Active Transport, Cell Nucleus , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , RNA, Small Interfering/genetics , Active Transport, Cell Nucleus/genetics , Benzamides/pharmacology , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Gene Knockdown Techniques , Gene Library , Humans , Hydrazines/pharmacology , Imatinib Mesylate , K562 Cells , Karyopherins/antagonists & inhibitors , Karyopherins/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Triazoles/pharmacology , Tumor Stem Cell Assay , ran GTP-Binding Protein/antagonists & inhibitors , ran GTP-Binding Protein/genetics , Exportin 1 Protein
11.
Leukemia ; 29(3): 586-597, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25134459

ABSTRACT

Mutations in the BCR-ABL1 kinase domain are an established mechanism of tyrosine kinase inhibitor (TKI) resistance in Philadelphia chromosome-positive leukemia, but fail to explain many cases of clinical TKI failure. In contrast, it is largely unknown why some patients fail TKI therapy despite continued suppression of BCR-ABL1 kinase activity, a situation termed BCR-ABL1 kinase-independent TKI resistance. Here, we identified activation of signal transducer and activator of transcription 3 (STAT3) by extrinsic or intrinsic mechanisms as an essential feature of BCR-ABL1 kinase-independent TKI resistance. By combining synthetic chemistry, in vitro reporter assays, and molecular dynamics-guided rational inhibitor design and high-throughput screening, we discovered BP-5-087, a potent and selective STAT3 SH2 domain inhibitor that reduces STAT3 phosphorylation and nuclear transactivation. Computational simulations, fluorescence polarization assays and hydrogen-deuterium exchange assays establish direct engagement of STAT3 by BP-5-087 and provide a high-resolution view of the STAT3 SH2 domain/BP-5-087 interface. In primary cells from chronic myeloid leukemia (CML) patients with BCR-ABL1 kinase-independent TKI resistance, BP-5-087 (1.0 µM) restored TKI sensitivity to therapy-resistant CML progenitor cells, including leukemic stem cells. Our findings implicate STAT3 as a critical signaling node in BCR-ABL1 kinase-independent TKI resistance, and suggest that BP-5-087 has clinical utility for treating malignancies characterized by STAT3 activation.


Subject(s)
Aminosalicylic Acids/pharmacology , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Leukocytes, Mononuclear/drug effects , Neoplastic Stem Cells/drug effects , STAT3 Transcription Factor/genetics , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Aminosalicylic Acids/chemical synthesis , Aminosalicylic Acids/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides/pharmacology , Cell Line, Tumor , Dasatinib , Drug Discovery , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Genes, Reporter , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Luciferases/genetics , Luciferases/metabolism , Molecular Docking Simulation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyrimidines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Signal Transduction , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Thiazoles/pharmacology
12.
Cancer Cell ; 26(3): 428-442, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25132497

ABSTRACT

Ponatinib is the only currently approved tyrosine kinase inhibitor (TKI) that suppresses all BCR-ABL1 single mutants in Philadelphia chromosome-positive (Ph(+)) leukemia, including the recalcitrant BCR-ABL1(T315I) mutant. However, emergence of compound mutations in a BCR-ABL1 allele may confer ponatinib resistance. We found that clinically reported BCR-ABL1 compound mutants center on 12 key positions and confer varying resistance to imatinib, nilotinib, dasatinib, ponatinib, rebastinib, and bosutinib. T315I-inclusive compound mutants confer high-level resistance to TKIs, including ponatinib. In vitro resistance profiling was predictive of treatment outcomes in Ph(+) leukemia patients. Structural explanations for compound mutation-based resistance were obtained through molecular dynamics simulations. Our findings demonstrate that BCR-ABL1 compound mutants confer different levels of TKI resistance, necessitating rational treatment selection to optimize clinical outcome.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Pyridazines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Catalytic Domain , Fusion Proteins, bcr-abl/chemistry , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Molecular Dynamics Simulation , Mutation, Missense , Philadelphia Chromosome , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridazines/chemistry , Pyridazines/therapeutic use , Treatment Failure
13.
Stem Cells ; 32(7): 1774-88, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24648383

ABSTRACT

The bone morphogenetic protein antagonist Gremlin 2 (Grem2) is required for atrial differentiation and establishment of cardiac rhythm during embryonic development. A human Grem2 variant has been associated with familial atrial fibrillation, suggesting that abnormal Grem2 activity causes arrhythmias. However, it is not known how Grem2 integrates into signaling pathways to direct atrial cardiomyocyte differentiation. Here, we demonstrate that Grem2 expression is induced concurrently with the emergence of cardiovascular progenitor cells during differentiation of mouse embryonic stem cells (ESCs). Grem2 exposure enhances the cardiogenic potential of ESCs by 20-120-fold, preferentially inducing genes expressed in atrial myocytes such as Myl7, Nppa, and Sarcolipin. We show that Grem2 acts upstream to upregulate proatrial transcription factors CoupTFII and Hey1 and downregulate atrial fate repressors Irx4 and Hey2. The molecular phenotype of Grem2-induced atrial cardiomyocytes was further supported by induction of ion channels encoded by Kcnj3, Kcnj5, and Cacna1d genes and establishment of atrial-like action potentials shown by electrophysiological recordings. We show that promotion of atrial-like cardiomyocytes is specific to the Gremlin subfamily of BMP antagonists. Grem2 proatrial differentiation activity is conveyed by noncanonical BMP signaling through phosphorylation of JNK and can be reversed by specific JNK inhibitors, but not by dorsomorphin, an inhibitor of canonical BMP signaling. Taken together, our data provide novel mechanistic insights into atrial cardiomyocyte differentiation from pluripotent stem cells and will assist the development of future approaches to study and treat arrhythmias.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , MAP Kinase Signaling System , Proteins/physiology , Animals , Cells, Cultured , Cytokines , Heart Atria/cytology , Mice , Myocytes, Cardiac/physiology
14.
Curr Protoc Pharmacol ; Chapter 14: Unit14.24, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23456612

ABSTRACT

Zebrafish models continue to gain popularity as in vivo models for drug discovery. Described in this overview are advantages and challenges of zebrafish drug screening, as well as a novel in vivo screen for immunomodulatory compounds using transgenic, T cell reporting zebrafish larvae designed for discovery of compounds targeting T cell leukemia. This assay system allows rapid screening of large numbers of compounds while avoiding the pitfalls of assays based on cell cultures, which lack biologic context and are afflicted by genomic instability. The rationale for this approach is based on similarities of immature normal T cells and developmentally arrested, malignant lymphoblasts in mammalian species. The screening algorithm has been used to identify a nontoxic compound with activity in both acute leukemia models and models of multiple sclerosis, demonstrating the utility of this screening procedure.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Lymphocyte Activation/drug effects , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/immunology , Zebrafish
15.
Bioorg Med Chem Lett ; 22(22): 6974-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23046966

ABSTRACT

AZD9272 and AZD6538 are two novel mGluR5 negative allosteric modulators selected for further clinical development. An initial high-throughput screening revealed leads with promising profiles, which were further optimized by minor, yet indispensable, structural modifications to bring forth these drug candidates. Advantageously, both compounds may be synthesized in as little as one step. Both are highly potent and selective for the human as well as the rat mGluR5 where they interact at the same binding site than MPEP. They are orally available, allow for long interval administration due to a high metabolic stability and long half-lives in rats and permeate the blood brain barrier to a high extent. AZD9272 has progressed into phase I clinical studies.


Subject(s)
Oxadiazoles/chemistry , Pyridines/chemistry , Receptors, Metabotropic Glutamate/chemistry , Allosteric Regulation , Animals , Binding Sites , Central Nervous System/diagnostic imaging , Drug Evaluation, Preclinical , HEK293 Cells , Half-Life , Humans , Isotope Labeling , Male , Microsomes/metabolism , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Radionuclide Imaging , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
16.
Immunohematology ; 27(3): 104-6, 2011.
Article in English | MEDLINE | ID: mdl-22356550

ABSTRACT

We analyzed our historic patient database at North Shore University Hospital and determined both the overall frequency of anti-Js(a) and the frequency at which it was detected in combination with other alloantibodies to red blood cell (RBC) antigens. Screening cells used currently are negative for Js(a). Our data suggest that anti-Js(a) would not be detected in 30 to 40 percent of patients in which it is the sole antibody present. Since 1996 the antibody was only detected when other antibodies were found in the screening process. We are exposing 1.7 percent of our patients (90 patients/year) to Js(a). The clinical significance of anti-Js(a) is apparent with previous literature, and our conclusion is that additional studies should be performed to determine whether Js(a) should be included in current antibody screening cells.


Subject(s)
Autoantibodies/blood , Blood Group Incompatibility/blood , Blood Grouping and Crossmatching , Kell Blood-Group System/blood , Blood Group Incompatibility/epidemiology , Databases, Bibliographic , Humans , New York City , Population Groups , Prevalence , Retrospective Studies
17.
Bioorg Med Chem Lett ; 15(10): 2537-40, 2005 May 16.
Article in English | MEDLINE | ID: mdl-15863312

ABSTRACT

Design, new synthesis, structure-activity relationship studies and calcium receptor antagonist (calcilytic) properties of novel 3H-pyrimidin-4-ones are described.


Subject(s)
Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacology , Receptors, Calcium-Sensing/drug effects , Pyrimidinones/chemistry , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 15(6): 1557-60, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15745796

ABSTRACT

Structure-activity relationship studies, focused on identification of the active pharmacophore fragments in a single high-throughput screening calcilytic hit, resulted in the discovery of potent calcium receptor antagonists, substituted 3H-quinazolin-4-ones.


Subject(s)
Quinazolines/chemistry , Quinazolines/pharmacology , Receptors, Calcium-Sensing/antagonists & inhibitors , Animals , Male , Models, Chemical , Molecular Structure , Osteoporosis/drug therapy , Parathyroid Hormone/blood , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
19.
J Pharmacol Exp Ther ; 308(2): 627-35, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14593085

ABSTRACT

Calcimimetic compounds, which activate the parathyroid cell Ca(2+) receptor (CaR) and inhibit parathyroid hormone (PTH) secretion, are under experimental study as a treatment for hyperparathyroidism. This report describes the salient pharmacodynamic properties, using several test systems, of a new calcimimetic compound, cinacalcet HCl. Cinacalcet HCl increased the concentration of cytoplasmic Ca(2+) ([Ca(2+)](i)) in human embryonic kidney 293 cells expressing the human parathyroid CaR. Cinacalcet HCl (EC(50) = 51 nM) in the presence of 0.5 mM extracellular Ca(2+) elicited increases in [Ca(2+)](i) in a dose- and calcium-dependent manner. Similarly, in the presence of 0.5 mM extracellular Ca(2+), cinacalcet HCl (IC(50) = 28 nM) produced a concentration-dependent decrease in PTH secretion from cultured bovine parathyroid cells. Using rat medullary thyroid carcinoma 6-23 cells expressing the CaR, cinacalcet HCl (EC(50) = 34 nM) produced a concentration-dependent increase in calcitonin secretion. In vivo studies in rats demonstrated cinacalcet HCl is orally bioavailable and displays approximately linear pharmacokinetics over the dose range of 1 to 36 mg/kg. Furthermore, this compound suppressed serum PTH and blood-ionized Ca(2+) levels and increased serum calcitonin levels in a dose-dependent manner. Cinacalcet was about 30-fold more potent at lowering serum levels of PTH than it was at increasing serum calcitonin levels. The S-enantiomer of cinacalcet (S-AMG 073) was at least 75-fold less active in these assay systems. The present findings provide compelling evidence that cinacalcet HCl is a potent and stereoselective activator of the parathyroid CaR and, as such, might be beneficial in the treatment of hyperparathyroidism.


Subject(s)
Calcitonin/metabolism , Naphthalenes/pharmacology , Parathyroid Glands/drug effects , Parathyroid Hormone/metabolism , Animals , Calcitonin/blood , Calcium/blood , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cinacalcet , Humans , Male , Naphthalenes/pharmacokinetics , Parathyroid Glands/metabolism , Parathyroid Hormone/blood , Phosphorus/blood , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...