Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Commun ; 12(1): 2431, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893273

ABSTRACT

Insulin resistance and lower muscle quality (strength divided by mass) are hallmarks of type 2 diabetes (T2D). Here, we explore whether alterations in muscle stem cells (myoblasts) from individuals with T2D contribute to these phenotypes. We identify VPS39 as an important regulator of myoblast differentiation and muscle glucose uptake, and VPS39 is downregulated in myoblasts and myotubes from individuals with T2D. We discover a pathway connecting VPS39-deficiency in human myoblasts to impaired autophagy, abnormal epigenetic reprogramming, dysregulation of myogenic regulators, and perturbed differentiation. VPS39 knockdown in human myoblasts has profound effects on autophagic flux, insulin signaling, epigenetic enzymes, DNA methylation and expression of myogenic regulators, and gene sets related to the cell cycle, muscle structure and apoptosis. These data mimic what is observed in myoblasts from individuals with T2D. Furthermore, the muscle of Vps39+/- mice display reduced glucose uptake and altered expression of genes regulating autophagy, epigenetic programming, and myogenesis. Overall, VPS39-deficiency contributes to impaired muscle differentiation and reduced glucose uptake. VPS39 thereby offers a therapeutic target for T2D.


Subject(s)
Autophagy-Related Proteins/genetics , Autophagy/genetics , Cell Differentiation/genetics , Diabetes Mellitus, Type 2/genetics , Epigenomics/methods , Myoblasts/metabolism , Stem Cells/metabolism , Vesicular Transport Proteins/genetics , Animals , Autophagy-Related Proteins/deficiency , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Epigenesis, Genetic/genetics , Female , Gene Expression Profiling/methods , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle Development/genetics , Vesicular Transport Proteins/deficiency
2.
Cancer Res ; 78(21): 6257-6267, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30135191

ABSTRACT

Activation of p53 by inhibitors of the p53-MDM2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. Here, we report distinct mechanisms by which the novel, potent, and selective inhibitor of the p53-MDM2 interaction HDM201 elicits therapeutic efficacy when applied at various doses and schedules. Continuous exposure of HDM201 led to induction of p21 and delayed accumulation of apoptotic cells. By comparison, high-dose pulses of HDM201 were associated with marked induction of PUMA and a rapid onset of apoptosis. shRNA screens identified PUMA as a mediator of the p53 response specifically in the pulsed regimen. Consistent with this, the single high-dose HDM201 regimen resulted in rapid and marked induction of PUMA expression and apoptosis together with downregulation of Bcl-xL in vivo Knockdown of Bcl-xL was identified as the top sensitizer to HDM201 in vitro, and Bcl-xL was enriched in relapsing tumors from mice treated with intermittent high doses of HDM201. These findings define a regimen-dependent mechanism by which disruption of MDM2-p53 elicits therapeutic efficacy when given with infrequent dosing. In an ongoing HDM201 trial, the observed exposure-response relationship indicates that the molecular mechanism elicited by pulse dosing is likely reproducible in patients. These data support the clinical comparison of daily and intermittent regimens of p53-MDM2 inhibitors.Significance: Pulsed high doses versus sustained low doses of the p53-MDM2 inhibitor HDM201 elicit a proapoptotic response from wild-type p53 cancer cells, offering guidance to current clinical trials with this and other drugs that exploit the activity of p53. Cancer Res; 78(21); 6257-67. ©2018 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Area Under Curve , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Screening Assays, Antitumor , Humans , Imidazoles/pharmacology , Kaplan-Meier Estimate , Maximum Tolerated Dose , Mice , Neoplasm Transplantation , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Small Interfering/metabolism , Time Factors , bcl-X Protein/metabolism
4.
Elife ; 42015 May 12.
Article in English | MEDLINE | ID: mdl-25965177

ABSTRACT

Biomarkers for patient selection are essential for the successful and rapid development of emerging targeted anti-cancer therapeutics. In this study, we report the discovery of a novel patient selection strategy for the p53-HDM2 inhibitor NVP-CGM097, currently under evaluation in clinical trials. By intersecting high-throughput cell line sensitivity data with genomic data, we have identified a gene expression signature consisting of 13 up-regulated genes that predicts for sensitivity to NVP-CGM097 in both cell lines and in patient-derived tumor xenograft models. Interestingly, these 13 genes are known p53 downstream target genes, suggesting that the identified gene signature reflects the presence of at least a partially activated p53 pathway in NVP-CGM097-sensitive tumors. Together, our findings provide evidence for the use of this newly identified predictive gene signature to refine the selection of patients with wild-type p53 tumors and increase the likelihood of response to treatment with p53-HDM2 inhibitors, such as NVP-CGM097.


Subject(s)
Biomarkers/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Isoquinolines/pharmacology , Neoplasms/drug therapy , Patient Selection , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Acta Neuropathol Commun ; 2: 165, 2014 Dec 14.
Article in English | MEDLINE | ID: mdl-25510661

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a primary progressive neurodegenerative disease characterised by neuronal loss of lower motor neurons (in the spinal cord and brainstem) and/or upper motor neurons (in the motor cortex) and subsequent denervation atrophy of skeletal muscle. AIM: A comprehensive examination of muscle pathology from a cohort of clinically confirmed ALS patients, including an investigation of inflammation, complement activation, and deposition of abnormal proteins in order to compare them with findings from an age-matched, control group. MATERIAL AND METHODS: 31 muscle biopsies from clinically confirmed ALS patients and 20 normal controls underwent a comprehensive protocol of histochemical and immunohistochemical stains, including HLA-ABC, C5b-9, p62, and TDP-43. RESULTS: Neurogenic changes were confirmed in 30/31 ALS cases. In one case, no neurogenic changes could be detected. Muscle fibre necrosis was seen in 5/31 cases and chronic mononuclear inflammatory cell infiltration in 5/31 (2 of them overlapped with those showing muscle necrosis). In four biopsies there was an increase in the proportion of cytochrome oxidase (COX) negative fibres (2-3%). p62 faintly stained cytoplasmic bodies in eight cases and none were immunoreactive to TDP-43. CONCLUSION: This large series of muscle biopsies from patients with ALS demonstrates neurogenic atrophy is a nearly uniform finding and that mild mitochondrial abnormalities and low-grade inflammation can be seen and do not rule out the diagnosis of ALS. These findings could lend support to the notion that ALS is a complex and heterogeneous disorder.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Mitochondria/immunology , Mitochondria/pathology , Muscle, Skeletal/pathology , Adult , Amyotrophic Lateral Sclerosis/blood , Biopsy , Cohort Studies , DNA-Binding Proteins/metabolism , Electron Transport Complex IV/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Skeletal/immunology , Necrosis , RNA-Binding Proteins/metabolism
6.
PLoS One ; 9(5): e97803, 2014.
Article in English | MEDLINE | ID: mdl-24841795

ABSTRACT

UNLABELLED: The neurite outgrowth inhibitor, Nogo-A, has been shown to be overexpressed in skeletal muscle in amyotrophic lateral sclerosis (ALS); it is both a potential biomarker and therapeutic target. We performed a double-blind, two-part, dose-escalation study, in subjects with ALS, assessing safety, pharmacokinetics (PK) and functional effects of ozanezumab, a humanized monoclonal antibody against Nogo-A. In Part 1, 40 subjects were randomized (3∶1) to receive single dose intravenous ozanezumab (0.01, 0.1, 1, 5, or 15 mg/kg) or placebo. In Part 2, 36 subjects were randomized (3∶1) to receive two repeat doses of intravenous ozanezumab (0.5, 2.5, or 15 mg/kg) or placebo, approximately 4 weeks apart. The primary endpoints were safety and tolerability (adverse events [AEs], vital signs, electrocardiogram (ECG), and clinical laboratory tests). Secondary endpoints included PK, immunogenicity, functional endpoints (clinical and electrophysiological), and biomarker parameters. Overall, ozanezumab treatment (0.01-15 mg/kg) was well tolerated. The overall incidence of AEs in the repeat dose 2.5 mg/kg and 15 mg/kg ozanezumab groups was higher than in the repeat dose placebo group and repeat dose 0.5 mg/kg ozanezumab group. The majority were considered not related to study drug by the investigators. Six serious AEs were reported in three subjects receiving ozanezumab; none were considered related to study drug. No study drug-related patterns were identified for ECG, laboratory, or vital signs parameters. One subject (repeat dose 15 mg/kg ozanezumab) showed a weak, positive anti-ozanezumab-antibody result. PK results were generally consistent with monoclonal antibody treatments. No apparent treatment effects were observed for functional endpoints or muscle biomarkers. Immunohistochemical staining showed dose-dependent co-localization of ozanezumab with Nogo-A in skeletal muscle. In conclusion, single and repeat dose ozanezumab treatment was well tolerated and demonstrated co-localization at the site of action. These findings support future studies with ozanezumab in ALS. TRIAL REGISTRATION: ClinicalTrials.gov NCT00875446 GSK-ClinicalStudyRegister.com GSK ID 111330.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Myelin Proteins/metabolism , Administration, Intravenous , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers/metabolism , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nogo Proteins
7.
Am J Nucl Med Mol Imaging ; 2(3): 260-70, 2012.
Article in English | MEDLINE | ID: mdl-23133816

ABSTRACT

2'-Fluoro-2'-deoxy-1ß-D-arabinofuranosyl-5-[(125)I]iodouracil ([(125)I]FIAU), a substrate for the thymidine kinase (TK) present in most bacteria, has been used as an imaging agent for single photon emission computed tomography (SPECT) in an experimental model of lung infection. Using SPECT-CT we show that [(125)I]FIAU is specific for bacterial infection rather than sterile inflammation. We report [(125)I]FIAU lung uptake values of 1.26 ± 0.20 percent injected dose per gram (%ID/g) in normal controls, 1.69 ± 0.32 %ID/g in lung inflammation and up to 7.14 ± 1.09 %ID/g in lung infection in ex vivo biodistribution studies at 24 h after intranasal administration of bacteria. Images of [(125)I]FIAU signal within lung can be used to estimate the number of bacteria present, with a limit of detection of 10(9) colony forming units per mL on the X-SPECT scanner. [(125)I]FIAU-Based bacterial imaging may be useful in preclinical models to facilitate the development of new antibiotics, particularly in cases where a corresponding human trial is planned.

8.
Clin Drug Investig ; 32(3): 179-87, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22217154

ABSTRACT

BACKGROUND AND OBJECTIVES: Severe sepsis and septic shock have posed significant treatment challenges for many years. Recently, a number of circulating apoptosis biomarkers have emerged, such as full-length and caspase-cleaved cytokeratin 18 (CK18) and nucleosomal DNA (nDNA), that may be predictive of likely outcome. This non-interventional study aimed to assess the ability of enzyme-linked immunosorbent assays (ELISAs) for these biomarkers to provide clinically useful information to guide the management of sepsis. METHODS: This study was conducted in patients admitted to the intensive care unit with severe sepsis at five US centres. Blood samples for assessment of plasma levels of full-length CK18 (measured by the M65® ELISA) and caspase-cleaved CK18 (measured by the M30-Apoptosense® ELISA) and nDNA (measured by ELISA) were collected from patients within 2 hours of consent (baseline) and on days 2, 4 and 8. Blood samples from 17 healthy volunteers acted as controls. Levels of each biomarker were presented descriptively. RESULTS: A total of 22 patients (mean age 60 [range, 24-83] years; 50% male) were included in the study. The mean APACHE II score was 24.4 (range 7-50). One-third of patients had three organ system failures and over one-half had septic shock. Three patients died during the study. Full-length and caspase-cleaved CK18 levels decreased within 48 hours following initiation of treatment of sepsis in patients who survived, whereas increases were observed in the same timeframe in patients who died within 28 days of admission. Baseline nDNA and total soluble CK18 levels (caspase-cleaved and total intact) were significantly (p ≤ 0.05) higher in patients who required renal support than those who did not. CONCLUSIONS: Despite the small numbers of subjects assessed in the current study, these results confirm that measurement of apoptosis biomarkers may help to provide clinically useful information to manage sepsis and expedite development of novel therapeutics. However, further investigations to fully assess their prognostic value are required.


Subject(s)
DNA/blood , Keratin-18/blood , Nucleosomes/chemistry , Sepsis/blood , APACHE , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Pilot Projects , Prognosis , Sepsis/mortality
9.
Immunobiology ; 216(3): 343-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20961651

ABSTRACT

The pleiotropic cytokine transforming growth factor-ß (TGF-ß) signals through different pathways among which the Smad- and the MAP-Kinase pathways are already well characterized. Both pathways utilize adaptor/chaperone molecules that facilitate or modulate the intracellular signaling events. Two of the proteins shown in vitro to play a role in Smad-dependent signaling are the TGF-ß Receptor Associated Protein-1 (TRAP1, also TGFBRAP1) and its homologue VPS39, also known as Vam6 and TRAP1-Like-Protein (TLP). We generated mice deficient for TRAP1 and VPS39/TLP, respectively. Absence of TRAP1 protein results in death at either of two defined timepoints during embryogenesis, before the blastula stage or during gastrulation, whereas most of the VPS39 deficient mice die before E6.5. Heterozygous mice show no overt phenotype. In summary, our data indicate that TRAP1 and VPS39 are nonredundant and essentially required for early embryonic development.


Subject(s)
Blastula/embryology , Embryonic Development , Gastrula/embryology , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Animals, Genetically Modified , Autophagy-Related Proteins , Blotting, Northern , Blotting, Western , Cells, Cultured , Gene Expression , Genotype , Guanine Nucleotide Exchange Factors/genetics , HSP90 Heat-Shock Proteins , Intracellular Signaling Peptides and Proteins/genetics , Mice , Polymerase Chain Reaction , Signal Transduction , Vesicular Transport Proteins
10.
Ann Clin Microbiol Antimicrob ; 9: 5, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20096100

ABSTRACT

BACKGROUND: Accurate measurement of the incidence of diarrhoeagenic E. coli in patients with diarrhoea is hindered by the current methods of detection and varies from country to country. In order to improve the diagnosis of diarrhoeagenic E. coli (DEC), we developed a set of multiplex TaqMan real-time PCRs designed to detect the respective pathogens from an overnight stool culture. METHODS: Over the period Jan. 2006 to Dec. 2006 all stool specimens (n = 1981) received were investigated for EPEC and EAEC. RESULTS: Of these, 371 specimens had no growth of Enterobacteriaceae. Of the remaining 1610 specimens 144 (8,9%) were positive for EPEC and 78 (4,8%) positive for EAEC. Among the EPEC positive stool specimens 28 (19,4%) were received from the tropical diseases unit, 49 (34%) from the paediatric dept. and 67 (46,5%) from the remainder of the wards. The EAEC were distributed as follows: 39 (50%) - tropical diseases, 19 (24,4%) -paediatrics and 20 (25,6%) other wards. Proportionately more EAEC and EPEC were found in children less than 3 years of age than other age groups. In only 22,2% of the detected EPEC and 23% of EAEC was the investigation requested by hospital staff. CONCLUSIONS: This is, to our knowledge, the first study using a multiplex TaqMan PCR for the successful detection of diarrhoeagenic E. coli. In conclusion, due to the high prevalence of DEC detected, investigation of EPEC and EAEC should be recommended as a routine diagnostic test for patients with infectious diarrhoea.


Subject(s)
Enteropathogenic Escherichia coli/isolation & purification , Escherichia coli Infections/diagnosis , Escherichia coli/isolation & purification , Polymerase Chain Reaction/methods , Adolescent , Adult , Aged , Child , Child, Preschool , DNA Primers , Enteropathogenic Escherichia coli/genetics , Escherichia coli/genetics , Escherichia coli Infections/epidemiology , Feces/microbiology , Germany/epidemiology , Hospitals, University , Humans , Infant , Infant, Newborn , Middle Aged , Prevalence , Sensitivity and Specificity
11.
J Invest Dermatol ; 127(11): 2596-604, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17625598

ABSTRACT

E. coli is a gram-negative bacterium rarely found on human skin. We investigated whether direct interaction of E. coli with keratinocytes might induce an innate immune response through recognition by pattern recognition receptors. The capacity of E. coli to activate innate immune responses and IL-8 induction was investigated. We found that E. coli significantly induced human S100A7 and S100A15 transcript abundance and IL-8 release in cultured primary human keratinocytes. S100A15 is a member of the S100 protein family with previously unknown function. E. coli induced effects could be inhibited by neutralizing Toll-like receptor 4 (TLR4) antibodies, suggesting that E. coli-induced IL-8 and S100A15 expression in human keratinocytes are TLR4 dependent. TLR4-/- mice lacked elevated mS100A15 expression after infection with E. coli in contrast to wild-type mice. In vitro, human S100A15 displayed antimicrobial activity against E. coli. Our findings suggest that E. coli modulates S100A15 and IL-8 expression of keratinocytes by recognition through TLR4.


Subject(s)
Escherichia coli/physiology , Keratinocytes/metabolism , S100 Proteins/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Female , Gene Expression Regulation/physiology , Humans , Immunity, Innate/immunology , Interleukin-8/genetics , Interleukin-8/metabolism , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , S100 Calcium Binding Protein A7 , S100 Proteins/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
12.
J Clin Immunol ; 27(6): 568-79, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17641957

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine that mainly acts as an inhibitor of immune functions. A lack of functional TGF-beta leads to autoimmune disease in animal models and dysregulated TGF-beta signaling is implicated in human autoimmune diseases. To define target genes that play a part in the inhibitory role of TGF-beta in the immune system, we have identified genes stimulated by TGF-beta in macrophages by gene-chip analysis. One of the TGF-beta regulated genes is carboxypeptidase D (CpD), a 180-kDa type I membrane protein. We have demonstrated that CpD is regulated by TGF-beta in various cell types of both, murine and human origin and, interestingly, is significantly downregulated in CD14 positive cells isolated from patients with lupus erythematosus (LE). Moreover, we show that downregulation of CpD leads to downmodulation of TGF-beta itself, suggesting a role for CpD in a positive feedback loop, providing further evidence for a role of this enzyme in LE. To our knowledge, this is the first report that demonstrates carboxypeptidase D as a TGF-beta target gene that is implicated in the pathogenesis of LE.


Subject(s)
Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/genetics , Gene Expression Regulation, Enzymologic/physiology , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/genetics , Transforming Growth Factor beta/physiology , Adult , Animals , Carboxypeptidases/biosynthesis , Cell Line , Cell Line, Tumor , Down-Regulation/genetics , Down-Regulation/immunology , Enzyme Induction/genetics , Enzyme Induction/immunology , Feedback, Physiological/immunology , Female , Humans , Lipopolysaccharide Receptors/metabolism , Lupus Erythematosus, Systemic/immunology , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology
14.
EMBO J ; 22(17): 4465-77, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12941698

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine signaling to the nucleus through cell surface transmembrane receptor serine/threonine kinases and cytoplasmic effectors, including Smad proteins. We describe a novel modulator of this pathway, TLP (TRAP-1-like protein), which is 25% identical to the previously described Smad4 chaperone, TRAP-1, and shows identical expression patterns in human tissues. Endogenous TLP associates with both active and kinase-deficient TGF-beta and activin type II receptors, but interacts with the common-mediator Smad4 only in the presence of TGF-beta/activin signaling. Overexpression of TLP represses the ability of TGF-beta to induce transcription from SBE-Luc, a Smad3/4-specific reporter, while it potentiates transcription from ARE-Luc, a Smad2/4-specific reporter. Consistent with this, TLP inhibits the formation of Smad3/4 complexes in the absence of effects on phosphorylation of Smad3, while it affects neither Smad2 phosphorylation nor hetero-oligomerization. We propose that TLP might regulate the balance of Smad2 and Smad3 signaling by localizing Smad4 intracellularly, thus contributing to cellular specificity of TGF-beta transcriptional responses in both normal and pathophysiology.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Trans-Activators/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Activins/metabolism , Amino Acid Sequence , Animals , Autophagy-Related Proteins , COS Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Endocytosis , Humans , Kinetics , Models, Biological , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Smad2 Protein , Smad3 Protein , Smad4 Protein , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection , Vesicular Transport Proteins
15.
Blood ; 101(2): 498-507, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12393416

ABSTRACT

We have investigated the role of Smad family proteins, known to be important cytoplasmic mediators of signals from the transforming growth factor-beta (TGF-beta) receptor serine/threonine kinases, in TGF-beta-dependent differentiation of hematopoietic cells, using as a model the human promyelocytic leukemia cell line, HL-60. TGF-beta-dependent differentiation of these cells to monocytes, but not retinoic acid-dependent differentiation to granulocytes, was accompanied by rapid phosphorylation and nuclear translocation of Smad2 and Smad3. Vitamin D(3) also induced phosphorylation of Smad2/3 and monocytic differentiation; however the effects were indirect, dependent on its ability to induce expression of TGF-beta1. Simultaneous treatment of these cells with TGF-beta1 and all-trans-retinoic acid (ATRA), which leads to almost equal numbers of granulocytes and monocytes, significantly reduced the level of phospho-Smad2/3 and its nuclear accumulation, compared with that in cells treated with TGF-beta1 alone. TGF-beta1 and ATRA activate P42/44 mitogen-activated protein (MAP) kinase with nearly identical kinetics, ruling out its involvement in these effects on Smad phosphorylation. Addition of the inhibitor-of-protein serine/threonine phosphatases, okadaic acid, blocks the ATRA-mediated reduction in TGF-beta-induced phospho-Smad2 and shifts the differentiation toward monocytic end points. In HL-60R mutant cells, which harbor a defective retinoic acid receptor-alpha (RAR-alpha), ATRA is unable to reduce levels of TGF-beta-induced phospho-Smad2/3, coincident with its inability to differentiate these cells along granulocytic pathways. Together, these data suggest a new level of cross-talk between ATRA and TGF-beta, whereby a putative RAR-alpha-dependent phosphatase activity limits the levels of phospho-Smad2/3 induced by TGF-beta, ultimately reducing the levels of nuclear Smad complexes mediating the TGF-beta-dependent differentiation of the cells to monocytic end points.


Subject(s)
DNA-Binding Proteins/physiology , Granulocytes/cytology , Monocytes/cytology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Cell Differentiation/drug effects , Cholecalciferol/pharmacology , DNA-Binding Proteins/metabolism , Drug Interactions , Granulocytes/drug effects , HL-60 Cells , Humans , Mitogen-Activated Protein Kinases/metabolism , Monocytes/drug effects , Phosphorylation/drug effects , Signal Transduction , Smad2 Protein , Smad3 Protein , Trans-Activators/metabolism , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...