62 results on '"Quelle FW"'
Search Results
2. Erythropoietin induces association of the JAK2 protein tyrosine kinase with the erythropoietin receptor in vivo
- Author
-
Miura, O, primary, Nakamura, N, additional, Quelle, FW, additional, Witthuhn, BA, additional, Ihle, JN, additional, and Aoki, N, additional
- Published
- 1994
- Full Text
- View/download PDF
3. High-level expression and purification of a recombinant human erythropoietin produced using a baculovirus vector
- Author
-
Quelle, FW, Caslake, LF, Burkert, RE, and Wojchowski, DM
- Abstract
Conditions presently have been established for the high-level expression and simplified purification of recombinant human erythropoietin produced in Spodoptera frugiperda cells. Expression, as mediated by infection with a recombinant baculovirus, was accomplished in suspension culture using reduced levels of serum and media supplements experimentally determined to provide optimum levels of factor production (500,000 U/L). Purification of this recombinant human erythropoietin to virtual homogeneity (greater than or equal to 99%) was accomplished via a simple three-step procedure involving isocratic elution from DEAE-Sephacel, reverse-phase high performance liquid chromatography (HPLC) on a C4 medium, and the single-step elution of purified hormone from concanavalin A agarose. Overall, an 890-fold purification was accomplished with a recovery of 80% as assayed in vitro. Biologically, this purified erythropoietin is highly active, possessing a specific activity in vitro of 200,000 U/mg protein. Chemically, this erythropoietin (molecular weight [mol wt] 26,200) appears exceptionally uniform in its oligosaccharide constitution (30%) as contrasted with heterogeneously glycosylated erythropoietins derived from mammalian cells (mol wt 30,000 to 38,000; 40% to 50% complex-type oligosaccharide). Thus, human erythropoietin as presently produced in an insect cell line comprises not only an abundant source of highly active, readily purified hormone for studies of its mechanism of action and cell surface receptor, but also represents a uniquely homogeneous form that should prove advantageous for direct structural analyses.
- Published
- 1989
- Full Text
- View/download PDF
4. Isolation and characterization of brain lipids by solid phase extraction and thin layer chromatography
- Author
-
Schweisguth, DC, primary, Quelle, FW, additional, Wachob, G, additional, and Hammerstedt, RH, additional
- Published
- 1989
- Full Text
- View/download PDF
5. Thermal Distortion of Diffraction-Limited Optical Elements
- Author
-
Quelle Fw
- Subjects
Diffraction ,education.field_of_study ,Materials science ,business.industry ,Materials Science (miscellaneous) ,Population ,Physics::Optics ,Joule ,Population inversion ,Laser ,Industrial and Manufacturing Engineering ,law.invention ,Optics ,law ,Optical cavity ,Thermal ,Business and International Management ,education ,business ,Refractive index - Abstract
Recent developments of laser materials have advanced the state of the art to a point where the optical quality of many of these materials is approaching the diffraction limit. Using such components in a laser does not necessarily guarantee the generation of diffraction-limited laser beams. One of the severe problems is thermal distortion introduced in the optical cavity by the flash lamps. Ruby and glass lasers require a minimum of 0.6 J of heat deposition per joule of population inversion. Typical figures are 4-6 J per joule of population inversion.(1) Nonuniformities in the deposition of this heat cause optical distortions which virtually preclude diffraction-limited laser operation even if the materials themselves are of diffraction-limited optical quality. This paper will investigate these thermal effects in detail, and the relative sensitivity of a variety of materials to nonuniform energy depositions will be discussed. Water close to its point of maximum density and a certain special type of glass known as Pockels glass will be shown to have properties of particular interest for use in diffraction-limited lasers.
- Published
- 1966
6. RhoBTB1 reverses established arterial stiffness in angiotensin II-induced hypertension by promoting actin depolymerization.
- Author
-
Fang S, Wu J, Reho JJ, Lu KT, Brozoski DT, Kumar G, Werthman AM, Silva SD Jr, Muskus Veitia PC, Wackman KK, Mathison AJ, Teng BQ, Lin CW, Quelle FW, and Sigmund CD
- Subjects
- Actins genetics, Actins metabolism, Angiotensin II metabolism, Animals, Mice, Muscle, Smooth, Vascular metabolism, Vascular Remodeling, Hypertension metabolism, Vascular Stiffness
- Abstract
Arterial stiffness predicts cardiovascular disease and all-cause mortality, but its treatment remains challenging. Mice treated with angiotensin II (Ang II) develop hypertension, arterial stiffness, vascular dysfunction, and a downregulation of Rho-related BTB domain-containing protein 1 (RhoBTB1) in the vasculature. RhoBTB1 is associated with blood pressure regulation, but its function is poorly understood. We tested the hypothesis that restoring RhoBTB1 can attenuate arterial stiffness, hypertension, and vascular dysfunction in Ang II-treated mice. Genetic complementation of RhoBTB1 in the vasculature was achieved using mice expressing a tamoxifen-inducible, smooth muscle-specific RhoBTB1 transgene. RhoBTB1 restoration efficiently and rapidly alleviated arterial stiffness but not hypertension or vascular dysfunction. Mechanistic studies revealed that RhoBTB1 had no substantial effect on several classical arterial stiffness contributors, such as collagen deposition, elastin content, and vascular smooth muscle remodeling. Instead, Ang II increased actin polymerization in the aorta, which was reversed by RhoBTB1. Changes in the levels of 2 regulators of actin polymerization, cofilin and vasodilator-stimulated phosphoprotein, in response to RhoBTB1 were consistent with an actin depolymerization mechanism. Our study reveals an important function of RhoBTB1, demonstrates its vital role in antagonizing established arterial stiffness, and further supports a functional and mechanistic separation among hypertension, vascular dysfunction, and arterial stiffness.
- Published
- 2022
- Full Text
- View/download PDF
7. Conditional deletion of smooth muscle Cullin-3 causes severe progressive hypertension.
- Author
-
Agbor LN, Nair AR, Wu J, Lu KT, Davis DR, Keen HL, Quelle FW, McCormick JA, Singer JD, and Sigmund CD
- Subjects
- Animals, Aorta metabolism, Aorta pathology, Cyclic GMP metabolism, Disease Models, Animal, Hypertension pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle, Smooth, Vascular metabolism, Mutation, Myocytes, Smooth Muscle metabolism, Nitric Oxide, Soluble Guanylyl Cyclase metabolism, Transcriptome, Vascular Stiffness, Vasodilation, Cullin Proteins genetics, Cullin Proteins metabolism, Genetic Predisposition to Disease genetics, Hypertension genetics, Hypertension metabolism, Muscle, Smooth metabolism
- Abstract
Patients with mutations in Cullin-3 (CUL3) exhibit severe early onset hypertension but the contribution of the smooth muscle remains unclear. Conditional genetic ablation of CUL3 in vascular smooth muscle (S-CUL3KO) causes progressive impairment in responsiveness to nitric oxide (NO), rapid development of severe hypertension, and increased arterial stiffness. Loss of CUL3 in primary aortic smooth muscle cells or aorta resulted in decreased expression of the NO receptor, soluble guanylate cyclase (sGC), causing a marked reduction in cGMP production and impaired vasodilation to cGMP analogues. Vasodilation responses to a selective large conductance Ca2+-activated K+-channel activator were normal suggesting that downstream signals which promote smooth muscle-dependent relaxation remained intact. We conclude that smooth muscle specific CUL3 ablation impairs both cGMP production and cGMP responses and that loss of CUL3 function selectively in smooth muscle is sufficient to cause severe hypertension by interfering with the NO-sGC-cGMP pathway. Our study provides compelling evidence for the sufficiency of vascular smooth muscle CUL3 as a major regulator of BP. CUL3 mutations cause severe vascular dysfunction, arterial stiffness and hypertension due to defects in vascular smooth muscle.
- Published
- 2019
- Full Text
- View/download PDF
8. RhoBTB1 protects against hypertension and arterial stiffness by restraining phosphodiesterase 5 activity.
- Author
-
Mukohda M, Fang S, Wu J, Agbor LN, Nair AR, Ibeawuchi SC, Hu C, Liu X, Lu KT, Guo DF, Davis DR, Keen HL, Quelle FW, and Sigmund CD
- Subjects
- Angiotensin II adverse effects, Angiotensin II pharmacology, Animals, Cullin Proteins genetics, Cullin Proteins metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 genetics, Disease Models, Animal, HEK293 Cells, Humans, Hypertension chemically induced, Hypertension genetics, Hypertension metabolism, Mice, Mice, Transgenic, Muscle, Smooth, Vascular pathology, Nitric Oxide genetics, Nitric Oxide metabolism, rho GTP-Binding Proteins genetics, rho-Associated Kinases genetics, rho-Associated Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Hypertension prevention & control, Muscle, Smooth, Vascular metabolism, Vascular Stiffness, Vasodilation, rho GTP-Binding Proteins metabolism
- Abstract
Mice selectively expressing PPARγ dominant negative mutation in vascular smooth muscle exhibit RhoBTB1-deficiency and hypertension. Our rationale was to employ genetic complementation to uncover the mechanism of action of RhoBTB1 in vascular smooth muscle. Inducible smooth muscle-specific restoration of RhoBTB1 fully corrected the hypertension and arterial stiffness by improving vasodilator function. Notably, the cardiovascular protection occurred despite preservation of increased agonist-mediated contraction and RhoA/Rho kinase activity, suggesting RhoBTB1 selectively controls vasodilation. RhoBTB1 augmented the cGMP response to nitric oxide by restraining the activity of phosphodiesterase 5 (PDE5) by acting as a substrate adaptor delivering PDE5 to the Cullin-3 E3 Ring ubiquitin ligase complex for ubiquitination inhibiting PDE5. Angiotensin-II infusion also caused RhoBTB1-deficiency and hypertension which was prevented by smooth muscle specific RhoBTB1 restoration. We conclude that RhoBTB1 protected from hypertension, vascular smooth muscle dysfunction, and arterial stiffness in at least two models of hypertension.
- Published
- 2019
- Full Text
- View/download PDF
9. RABL6A inhibits tumor-suppressive PP2A/AKT signaling to drive pancreatic neuroendocrine tumor growth.
- Author
-
Umesalma S, Kaemmer CA, Kohlmeyer JL, Letney B, Schab AM, Reilly JA, Sheehy RM, Hagen J, Tiwari N, Zhan F, Leidinger MR, O'Dorisio TM, Dillon J, Merrill RA, Meyerholz DK, Perl AL, Brown BJ, Braun TA, Scott AT, Ginader T, Taghiyev AF, Zamba GK, Howe JR, Strack S, Bellizzi AM, Narla G, Darbro BW, Quelle FW, and Quelle DE
- Subjects
- Carcinoma, Neuroendocrine genetics, Carcinoma, Neuroendocrine pathology, Cell Line, Tumor, Enzyme Activators pharmacology, G1 Phase drug effects, G1 Phase genetics, Humans, Oncogene Proteins genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Protein Phosphatase 2 genetics, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins c-akt genetics, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins genetics, rab GTP-Binding Proteins genetics, Carcinoma, Neuroendocrine enzymology, Oncogene Proteins metabolism, Pancreatic Neoplasms enzymology, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Hyperactivated AKT/mTOR signaling is a hallmark of pancreatic neuroendocrine tumors (PNETs). Drugs targeting this pathway are used clinically, but tumor resistance invariably develops. A better understanding of factors regulating AKT/mTOR signaling and PNET pathogenesis is needed to improve current therapies. We discovered that RABL6A, a new oncogenic driver of PNET proliferation, is required for AKT activity. Silencing RABL6A caused PNET cell-cycle arrest that coincided with selective loss of AKT-S473 (not T308) phosphorylation and AKT/mTOR inactivation. Restoration of AKT phosphorylation rescued the G1 phase block triggered by RABL6A silencing. Mechanistically, loss of AKT-S473 phosphorylation in RABL6A-depleted cells was the result of increased protein phosphatase 2A (PP2A) activity. Inhibition of PP2A restored phosphorylation of AKT-S473 in RABL6A-depleted cells, whereas PP2A reactivation using a specific small-molecule activator of PP2A (SMAP) abolished that phosphorylation. Moreover, SMAP treatment effectively killed PNET cells in a RABL6A-dependent manner and suppressed PNET growth in vivo. The present work identifies RABL6A as a new inhibitor of the PP2A tumor suppressor and an essential activator of AKT in PNET cells. Our findings offer what we believe is a novel strategy of PP2A reactivation for treatment of PNETs as well as other human cancers driven by RABL6A overexpression and PP2A inactivation.
- Published
- 2019
- Full Text
- View/download PDF
10. PPARγ and retinol binding protein 7 form a regulatory hub promoting antioxidant properties of the endothelium.
- Author
-
Woll AW, Quelle FW, and Sigmund CD
- Subjects
- Animals, Humans, Models, Biological, Protein Binding, Antioxidants metabolism, Endothelium, Vascular metabolism, PPAR gamma metabolism, Retinol-Binding Proteins, Cellular metabolism
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are a family of conserved ligand-activated nuclear receptor transcription factors heterogeneously expressed in mammalian tissues. PPARγ is recognized as a master regulator of adipogenesis, fatty acid metabolism, and glucose homeostasis, but genetic evidence also supports the concept that PPARγ regulates the cardiovascular system, particularly vascular function and blood pressure. There is now compelling evidence that the beneficial blood pressure-lowering effects of PPARγ activation are due to its activity in vascular smooth muscle and endothelium, through its modulation of nitric oxide-dependent vasomotor function. Endothelial PPARγ regulates the production and bioavailability of nitric oxide, while PPARγ in the smooth muscle regulates the vasomotor response to nitric oxide. We recently identified retinol binding protein 7 (RBP7) as a PPARγ target gene that is specifically and selectively expressed in the endothelium. In this review, we will discuss the evidence that RBP7 is required to mediate the antioxidant effects of PPARγ and mediate PPARγ target gene selectivity in the endothelium., (Copyright © 2017 the American Physiological Society.)
- Published
- 2017
- Full Text
- View/download PDF
11. Hypertension-Causing Mutation in Peroxisome Proliferator-Activated Receptor γ Impairs Nuclear Export of Nuclear Factor-κB p65 in Vascular Smooth Muscle.
- Author
-
Mukohda M, Lu KT, Guo DF, Wu J, Keen HL, Liu X, Ketsawatsomkron P, Stump M, Rahmouni K, Quelle FW, and Sigmund CD
- Subjects
- Active Transport, Cell Nucleus drug effects, Animals, Anti-Inflammatory Agents pharmacology, Cell Nucleus metabolism, Cells, Cultured, Fatty Acids, Unsaturated pharmacology, Inflammation genetics, Inflammation metabolism, Mice, Mutation, Tumor Necrosis Factor-alpha metabolism, Hypertension genetics, Hypertension metabolism, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular physiopathology, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, PPAR gamma genetics, Transcription Factor RelA metabolism
- Abstract
Selective expression of dominant negative (DN) peroxisome proliferator-activated receptor γ (PPARγ) in vascular smooth muscle cells (SMC) results in hypertension, atherosclerosis, and increased nuclear factor-κB (NF-κB) target gene expression. Mesenteric SMC were cultured from mice designed to conditionally express wild-type (WT) or DN-PPARγ in response to Cre recombinase to determine how SMC PPARγ regulates expression of NF-κB target inflammatory genes. SMC-specific overexpression of WT-PPARγ or agonist-induced activation of endogenous PPARγ blunted tumor necrosis factor α (TNF-α)-induced NF-κB target gene expression and activity of an NF-κB-responsive promoter. TNF-α-induced gene expression responses were enhanced by DN-PPARγ in SMC. Although expression of NF-κB p65 was unchanged, nuclear export of p65 was accelerated by WT-PPARγ and prevented by DN-PPARγ in SMC. Leptomycin B, a nuclear export inhibitor, blocked p65 nuclear export and inhibited the anti-inflammatory action of PPARγ. Consistent with a role in facilitating p65 nuclear export, WT-PPARγ coimmunoprecipitated with p65, and WT-PPARγ was also exported from the nucleus after TNF-α treatment. Conversely, DN-PPARγ does not bind to p65 and was retained in the nucleus after TNF-α treatment. Transgenic mice expressing WT-PPARγ or DN-PPARγ specifically in SMC (S-WT or S-DN) were bred with mice expressing luciferase controlled by an NF-κB-responsive promoter to assess effects on NF-κB activity in whole tissue. TNF-α-induced NF-κB activity was decreased in aorta and carotid artery from S-WT but was increased in vessels from S-DN mice. We conclude that SMC PPARγ blunts expression of proinflammatory genes by inhibition of NF-κB activity through a mechanism promoting nuclear export of p65, which is abolished by DN mutation in PPARγ., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
12. Retinol-binding protein 7 is an endothelium-specific PPAR γ cofactor mediating an antioxidant response through adiponectin.
- Author
-
Hu C, Keen HL, Lu KT, Liu X, Wu J, Davis DR, Ibeawuchi SC, Vogel S, Quelle FW, and Sigmund CD
- Subjects
- Adiponectin genetics, Animals, Diet, High-Fat, Endothelium, Vascular physiopathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Oxidative Stress, PPAR gamma genetics, RNA, Messenger genetics, Retinol-Binding Proteins, Cellular genetics, Adiponectin metabolism, Antioxidants metabolism, Endothelium, Vascular metabolism, PPAR gamma metabolism, Retinol-Binding Proteins, Cellular metabolism
- Abstract
Impaired PPARγ activity in endothelial cells causes oxidative stress and endothelial dysfunction which causes a predisposition to hypertension, but the identity of key PPARγ target genes that protect the endothelium remain unclear. Retinol-binding protein 7 (RBP7) is a PPARγ target gene that is essentially endothelium specific. Whereas RBP7-deficient mice exhibit normal endothelial function at baseline, they exhibit severe endothelial dysfunction in response to cardiovascular stressors, including high-fat diet and subpressor angiotensin II. Endothelial dysfunction was not due to differences in weight gain, impaired glucose homeostasis, or hepatosteatosis, but occurred through an oxidative stress-dependent mechanism which can be rescued by scavengers of superoxide. RNA sequencing revealed that RBP7 was required to mediate induction of a subset of PPARγ target genes by rosiglitazone in the endothelium including adiponectin. Adiponectin was selectively induced in the endothelium of control mice by high-fat diet and rosiglitazone, whereas RBP7 deficiency abolished this induction. Adiponectin inhibition caused endothelial dysfunction in control vessels, whereas adiponectin treatment of RBP7-deficient vessels improved endothelium-dependent relaxation and reduced oxidative stress. We conclude that RBP7 is required to mediate the protective effects of PPARγ in the endothelium through adiponectin, and RBP7 is an endothelium-specific PPARγ target and regulator of PPARγ activity., Competing Interests: Conflict of interest: The authors have declared that no conflict of interest exists.
- Published
- 2017
- Full Text
- View/download PDF
13. Cullin-3 mutation causes arterial stiffness and hypertension through a vascular smooth muscle mechanism.
- Author
-
Agbor LN, Ibeawuchi SC, Hu C, Wu J, Davis DR, Keen HL, Quelle FW, and Sigmund CD
- Subjects
- Animals, Cells, Cultured, Humans, Male, Mice, Mice, Inbred C57BL, Mutation, Myocytes, Smooth Muscle metabolism, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein metabolism, Cullin Proteins genetics, Hypertension genetics, Muscle, Smooth, Vascular physiopathology, Vascular Stiffness
- Abstract
Cullin-3 ( CUL3 ) mutations ( CUL3 Δ 9 ) were previously identified in hypertensive patients with pseudohypoaldosteronism type-II (PHAII), but the mechanism causing hypertension and whether this is driven by renal tubular or extratubular mechanisms remains unknown. We report that selective expression of CUL3Δ9 in smooth muscle acts by interfering with expression and function of endogenous CUL3, resulting in impaired turnover of the CUL3 substrate RhoA, increased RhoA activity, and augmented RhoA/Rho kinase signaling. This caused vascular dysfunction and increased arterial pressure under baseline conditions and a marked increase in arterial pressure, collagen deposition, and vascular stiffness in response to a subpressor dose of angiotensin II, which did not cause hypertension in control mice. Inhibition of total cullin activity increased the level of CUL3 substrates cyclin E and RhoA, and expression of CUL3Δ9 decreased the level of the active form of endogenous CUL3 in human aortic smooth muscle cells. These data indicate that selective expression of the Cul3 Δ 9 mutation in vascular smooth muscle phenocopies the hypertension observed in Cul3 Δ 9 human subjects and suggest that mutations in CUL3 cause human hypertension in part through a mechanism involving smooth muscle dysfunction initiated by a loss of CUL3-mediated degradation of RhoA., Competing Interests: The authors have declared that no conflict of interest exists.
- Published
- 2016
- Full Text
- View/download PDF
14. Endothelial PPAR-γ provides vascular protection from IL-1β-induced oxidative stress.
- Author
-
Mukohda M, Stump M, Ketsawatsomkron P, Hu C, Quelle FW, and Sigmund CD
- Subjects
- Animals, Antioxidants pharmacology, Aorta metabolism, Aorta pathology, Aorta physiopathology, Aortic Diseases metabolism, Aortic Diseases pathology, Aortic Diseases physiopathology, Dose-Response Relationship, Drug, Endothelial Cells metabolism, Endothelial Cells pathology, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Endothelium, Vascular physiopathology, Female, Gene Expression Regulation, Genotype, Humans, I-kappa B Proteins metabolism, Inflammation Mediators metabolism, Male, Mice, Inbred C57BL, Mice, Transgenic, NF-KappaB Inhibitor alpha, Nitric Oxide Synthase Type III metabolism, PPAR gamma agonists, PPAR gamma genetics, Phenotype, Phosphorylation, Reactive Oxygen Species metabolism, Signal Transduction drug effects, Transcription Factor RelA metabolism, Vasodilation drug effects, Vasodilator Agents pharmacology, Aorta drug effects, Aortic Diseases prevention & control, Endothelial Cells drug effects, Endothelium, Vascular drug effects, Interleukin-1beta pharmacology, Oxidative Stress drug effects, PPAR gamma metabolism
- Abstract
Loss of peroxisome proliferator-activated receptor (PPAR)-γ function in the vascular endothelium enhances atherosclerosis and NF-κB target gene expression in high-fat diet-fed apolipoprotein E-deficient mice. The mechanisms by which endothelial PPAR-γ regulates inflammatory responses and protects against atherosclerosis remain unclear. To assess functional interactions between PPAR-γ and inflammation, we used a model of IL-1β-induced aortic dysfunction in transgenic mice with endothelium-specific overexpression of either wild-type (E-WT) or dominant negative PPAR-γ (E-V290M). IL-1β dose dependently decreased IκB-α, increased phospho-p65, and increased luciferase activity in the aorta of NF-κB-LUC transgenic mice. IL-1β also dose dependently reduced endothelial-dependent relaxation by ACh. The loss of ACh responsiveness was partially improved by pretreatment of the vessels with the PPAR-γ agonist rosiglitazone or in E-WT. Conversely, IL-1β-induced endothelial dysfunction was worsened in the aorta from E-V290M mice. Although IL-1β increased the expression of NF-κB target genes, NF-κB p65 inhibitor did not alleviate endothelial dysfunction induced by IL-1β. Tempol, a SOD mimetic, partially restored ACh responsiveness in the IL-1β-treated aorta. Notably, tempol only modestly improved protection in the E-WT aorta but had an increased protective effect in the E-V290M aorta compared with the aorta from nontransgenic mice, suggesting that PPAR-γ-mediated protection involves antioxidant effects. IL-1β increased ROS and decreased the phospho-endothelial nitric oxide synthase (Ser(1177))-to-endothelial nitric oxide synthase ratio in the nontransgenic aorta. These effects were completely abolished in the aorta with endothelial overexpression of WT PPAR-γ but were worsened in the aorta with E-V290M even in the absence of IL-1β. We conclude that PPAR-γ protects against IL-1β-mediated endothelial dysfunction through a reduction of oxidative stress responses but not by blunting IL-1β-mediated NF-κB activity., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
15. Hypertension-causing Mutations in Cullin3 Protein Impair RhoA Protein Ubiquitination and Augment the Association with Substrate Adaptors.
- Author
-
Ibeawuchi SR, Agbor LN, Quelle FW, and Sigmund CD
- Subjects
- Carrier Proteins, Gene Expression, HEK293 Cells, Humans, Hypertension enzymology, Protein Binding, Protein Multimerization, Cullin Proteins genetics, Hypertension genetics, Ubiquitination, rhoA GTP-Binding Protein metabolism
- Abstract
Cullin-Ring ubiquitin ligases regulate protein turnover by promoting the ubiquitination of substrate proteins, targeting them for proteasomal degradation. It has been shown previously that mutations in Cullin3 (Cul3) causing deletion of 57 amino acids encoded by exon 9 (Cul3Δ9) cause hypertension. Moreover, RhoA activity contributes to vascular constriction and hypertension. We show that ubiquitination and degradation of RhoA is dependent on Cul3 in HEK293T cells in which Cul3 expression is ablated by either siRNA or by CRISPR-Cas9 genome editing. The latter was used to generate a Cul3-null cell line (HEK293T(Cul3KO)). When expressed in these cells, Cul3Δ9 supported reduced ubiquitin ligase activity toward RhoA compared with equivalent levels of wild-type Cul3 (Cul3WT). Consistent with its reduced activity, binding of Cul3Δ9 to the E3 ubiquitin ligase Rbx1 and neddylation of Cul3Δ9 were impaired significantly compared with Cul3WT. Conversely, Cul3Δ9 bound to substrate adaptor proteins more efficiently than Cul3WT. Cul3Δ9 also forms unstable dimers with Cul3WT, disrupting dimers of Cul3WT complexes that are required for efficient ubiquitination of some substrates. Indeed, coexpression of Cul3WT and Cul3Δ9 in HEK293T(Cul3KO) cells resulted in a decrease in the active form of Cul3WT. We conclude that Cul3Δ9-associated ubiquitin ligase activity toward RhoA is impaired and suggest that Cul3Δ9 mutations may act dominantly by sequestering substrate adaptors and disrupting Cul3WT complexes., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
16. RABL6A promotes G1-S phase progression and pancreatic neuroendocrine tumor cell proliferation in an Rb1-dependent manner.
- Author
-
Hagen J, Muniz VP, Falls KC, Reed SM, Taghiyev AF, Quelle FW, Gourronc FA, Klingelhutz AJ, Major HJ, Askeland RW, Sherman SK, O'Dorisio TM, Bellizzi AM, Howe JR, Darbro BW, and Quelle DE
- Subjects
- Cell Line, Tumor, Humans, Mitosis, Cell Proliferation, G1 Phase, Neuroendocrine Tumors pathology, Oncogene Proteins physiology, Pancreatic Neoplasms pathology, Retinoblastoma Protein physiology, S Phase, rab GTP-Binding Proteins physiology
- Abstract
Mechanisms of neuroendocrine tumor (NET) proliferation are poorly understood, and therapies that effectively control NET progression and metastatic disease are limited. We found amplification of a putative oncogene, RABL6A, in primary human pancreatic NETs (PNET) that correlated with high-level RABL6A protein expression. Consistent with those results, stable silencing of RABL6A in cultured BON-1 PNET cells revealed that it is essential for their proliferation and survival. Cells lacking RABL6A predominantly arrested in G1 phase with a moderate mitotic block. Pathway analysis of microarray data suggested activation of the p53 and retinoblastoma (Rb1) tumor-suppressor pathways in the arrested cells. Loss of p53 had no effect on the RABL6A knockdown phenotype, indicating that RABL6A functions independent of p53 in this setting. By comparison, Rb1 inactivation partially restored G1 to S phase progression in RABL6A-knockdown cells, although it was insufficient to override the mitotic arrest and cell death caused by RABL6A loss. Thus, RABL6A promotes G1 progression in PNET cells by inactivating Rb1, an established suppressor of PNET proliferation and development. This work identifies RABL6A as a novel negative regulator of Rb1 that is essential for PNET proliferation and survival. We suggest RABL6A is a new potential biomarker and target for anticancer therapy in PNET patients., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
17. ARF sees Pdgfrβ through the miR.
- Author
-
Reed SM, Quelle FW, and Quelle DE
- Subjects
- Animals, Cell Cycle Checkpoints genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, MicroRNAs metabolism
- Published
- 2014
- Full Text
- View/download PDF
18. Nuclear interactor of ARF and Mdm2 regulates multiple pathways to activate p53.
- Author
-
Reed SM, Hagen J, Tompkins VS, Thies K, Quelle FW, and Quelle DE
- Subjects
- Acetylation, Animals, Cell Line, Cell Line, Tumor, Cell Proliferation physiology, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Histone Acetyltransferases metabolism, Humans, Lysine Acetyltransferase 5, Mice, Protein Binding, Protein Structure, Tertiary, Proto-Oncogene Proteins c-mdm2 metabolism, Transcriptional Activation, Ubiquitination, Intracellular Signaling Peptides and Proteins metabolism, Nuclear Proteins metabolism, Signal Transduction, Tumor Suppressor Protein p53 metabolism
- Abstract
The p53 tumor suppressor is controlled by an interactive network of factors that stimulate or inhibit its transcriptional activity. Within that network, Mdm2 functions as the major antagonist of p53 by promoting its ubiquitylation and degradation. Conversely, Tip60 activates p53 through direct association on target promoters as well as acetylation of p53 at lysine 120 (K120). This study examines the functional relationship between Mdm2 and Tip60 with a novel p53 regulator, NIAM (nuclear interactor of ARF and Mdm2). Previous work showed NIAM can suppress proliferation and activate p53 independently of ARF, indicating that other factors mediate those activities. Here, we demonstrate that NIAM is a chromatin-associated protein that binds Tip60. NIAM can promote p53 K120 acetylation, although that modification is not required for NIAM to inhibit proliferation or induce p53 transactivation of the p21 promoter. Notably, Tip60 silencing showed it contributes to but is not sufficient for NIAM-mediated p53 activation, suggesting other mechanisms are involved. Indeed, growth-inhibitory forms of NIAM also bind to Mdm2, and increased NIAM expression levels disrupt p53-Mdm2 association, inhibit p53 polyubiquitylation, and prevent Mdm2-mediated inhibition of p53 transcriptional activity. Importantly, loss of NIAM significantly impairs p53 activation. Together, these results show that NIAM activates p53 through multiple mechanisms involving Tip60 association and Mdm2 inhibition. Thus, NIAM regulates 2 critical pathways that control p53 function and are altered in human cancers, implying an important role for NIAM in tumorigenesis.
- Published
- 2014
- Full Text
- View/download PDF
19. PPARγ: no SirT, no service.
- Author
-
Quelle FW and Sigmund CD
- Published
- 2013
- Full Text
- View/download PDF
20. FOXO transcription factors enforce cell cycle checkpoints and promote survival of hematopoietic cells after DNA damage.
- Author
-
Lei H and Quelle FW
- Subjects
- Animals, Cell Death drug effects, Cell Death radiation effects, Cell Line, Cell Proliferation drug effects, Cell Proliferation radiation effects, Cell Survival drug effects, Cell Survival radiation effects, Cisplatin pharmacology, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Drug Resistance drug effects, Drug Resistance radiation effects, Gamma Rays, Gene Knockdown Techniques, Hematopoietic System drug effects, Hematopoietic System radiation effects, Mice, Phosphoinositide-3 Kinase Inhibitors, Cell Cycle drug effects, Cell Cycle radiation effects, DNA Damage, Forkhead Transcription Factors metabolism, Hematopoietic System cytology, Hematopoietic System metabolism
- Abstract
The PI3K/AKT signaling pathway contributes to cell cycle progression of cytokine-dependent hematopoietic cells under normal conditions, and it is absolutely required to override DNA damage-induced cell cycle arrest checkpoints in these cells. Phosphatidylinositol-3-kinase (PI3K)/AKT activity also correlates with Cdk2 activity in hematopoietic cells, suggesting that Cdk2 activation may be a relevant end point for this signaling pathway. However, mediators downstream of AKT in this pathway have not been defined. The forkhead transcription factor O (FOXO) family are negatively regulated by AKT-dependent phosphorylation and are known regulators of genes affecting cell cycle progression. We show that enhanced FOXO activity replicates the effect of PI3K inhibitors in enforcing G(1) and G(2) phase arrest after DNA damage. Conversely, knockdown of endogenous FOXO proteins increased Cdk2 activity and overrode DNA damage checkpoints in cells lacking PI3K activity. Moreover, loss of FOXO activity caused an increase in sensitivity to cisplatin-induced cell death, which was associated with failure to arrest cell cycle progression in the face of DNA damage caused by this chemotherapeutic agent. These cell cycle arrests were dependent on p27 expression when mediated by FOXO3a alone, but also involve p27-independent mechanisms when promoted by endogenous FOXO proteins. Together, these observations show that FOXO proteins enforce DNA damage-induced cell cycle arrest in hematopoietic cells. Inhibition of FOXO activity by cytokine-induced PI3K/AKT signaling is sufficient to override these DNA damage-induced cell cycle checkpoints, but may negatively impact hematopoietic cell viability.
- Published
- 2009
- Full Text
- View/download PDF
21. Phosphoinositide 3-kinase signaling overrides a G2 phase arrest checkpoint and promotes aberrant cell cycling and death of hematopoietic cells after DNA damage.
- Author
-
Nimbalkar D and Quelle FW
- Subjects
- Animals, Cell Cycle Proteins metabolism, Cell Death radiation effects, Cell Line, Gamma Rays, Hematopoietic System radiation effects, Humans, Metaphase radiation effects, Mice, DNA Damage, G2 Phase radiation effects, Hematopoietic System cytology, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction radiation effects
- Abstract
DNA damage activates arrest checkpoints to halt cell cycle progression in G(1) and G(2) phases. These checkpoints can be overridden in hematopoietic cells by cytokines, such as erythropoietin, through the activation of a phosphoinositide 3-kinase (PI3K) signaling pathway. Here, we show that PI3K activity specifically overrides delayed mechanisms effecting permanent G(1) and G(2) phase arrests, but does not affect transient checkpoints arresting cells up to 10 hours after gamma-irradiation. Assessing the status of cell cycle regulators in hematopoietic cells arrested after gamma-irradiation, we show that Cdk2 activity is completely inhibited in both G(1) and G(2) arrested cells. Despite the absence of Cdk2 activity, cells arrested in G(2) phase did retain detectable levels of Cdk1 activity in the absence of PI3K signaling. However, reactivation of PI3K promoted robust increases in both Cdk1 and Cdk2 activity in G(2)-arrested cells. Reactivation of Cdks was accompanied by a resumption of cell cycling, but with strikingly different effectiveness in G(1) and G(2) phase arrested cells. Specifically, G(1)-arrested cells resumed normal cell cycle progression with little loss in viability when PI3K was activated after gamma-irradiation. Conversely, PI3K activation in G(2)-arrested cells promoted endoreduplication and death of the entire population. These observations show that cytokine-induced PI3K signaling pathways promote Cdk activation and override permanent cell cycle arrest checkpoints in hematopoietic cells. While this activity can rescue irradiated cells from permanent G(1) phase arrest, it results in aberrant cell cycling and death when activated in hematopoietic cells arrested at the G(2) phase DNA damage checkpoint.
- Published
- 2008
- Full Text
- View/download PDF
22. Cytokine signaling to the cell cycle.
- Author
-
Quelle FW
- Subjects
- Animals, Cell Proliferation, Humans, Lymphocytes cytology, Lymphocytes metabolism, Metabolic Networks and Pathways, Mitogen-Activated Protein Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, STAT Transcription Factors metabolism, Cell Cycle physiology, Cytokines metabolism, Intracellular Signaling Peptides and Proteins metabolism, Receptors, Cytokine metabolism, Signal Transduction
- Abstract
The proliferation of many myeloid and lymphoid cell populations is directly controlled by cytokine growth factors acting through a related family of cytokine receptors. This regulation implies that the signaling pathways activated by cytokine receptors must communicate with mechanisms that control mammalian cell cycle progression. Evidence for how these signaling pathways promote hematopoietic cell proliferation is considered along with their likely targets among the cell cycle regulators.
- Published
- 2007
- Full Text
- View/download PDF
23. Prolactin and heregulin override DNA damage-induced growth arrest and promote phosphatidylinositol-3 kinase-dependent proliferation in breast cancer cells.
- Author
-
Chakravarti P, Henry MK, and Quelle FW
- Subjects
- Blotting, Western, Cell Cycle, Cell Line, Tumor, Cell Proliferation, Cell Survival, Chromones pharmacology, DNA metabolism, Growth Substances, Humans, Morpholines pharmacology, Phosphorylation, Signal Transduction, Breast Neoplasms metabolism, DNA Damage, Gene Expression Regulation, Neoplastic, Neuregulin-1 physiology, Phosphatidylinositol 3-Kinases metabolism, Prolactin physiology
- Abstract
Heregulin (HRG), a ligand of ErbB receptor tyrosine kinases, is a potent mitogenic factor for breast cancer cells. Prolactin (PRL) has also been reported to regulate proliferation in breast cancer cells through its receptor, a member of the type I cytokine receptor family. Cytokine receptors are potent mitogens in hematopoietic cells, where they also override DNA damage-induced growth arrest checkpoints through activation of a phosphatidylinositol-3 kinase (PI3K) signaling pathway. In this study, we assessed the effect of gamma-irradiation on the mitogenic activity of HRG and PRL in breast cancer cells. HRG and PRL enhanced the proliferation of non-irradiated breast cancer cell lines in association with their ability to activate PI3K signaling pathways. Both growth factors also overrode irradiation-induced growth arrest in T47D cells, which resulted in decreased viability after irradiation. An inhibitor of PI3K, LY294002, abrogated growth factor-induced proliferation and the activity of cell cycle-dependent kinases in non-irradiated and irradiated cells. Thus, growth factors acting through distinct receptor families share a similar PI3K-dependent ability to promote proliferation and override DNA damage-induced growth arrest in breast cancer cells. These observations also suggest that selective activation of PI3K-dependent signaling can enhance radiosensitivity in breast cancer cells.
- Published
- 2005
24. Nucleophosmin (B23) targets ARF to nucleoli and inhibits its function.
- Author
-
Korgaonkar C, Hagen J, Tompkins V, Frazier AA, Allamargot C, Quelle FW, and Quelle DE
- Subjects
- Animals, COS Cells, Cell Nucleolus genetics, Cell Proliferation, Chlorocebus aethiops, Cyclin-Dependent Kinase Inhibitor p16, Humans, Mice, NIH 3T3 Cells, Nucleophosmin, Protein Binding, Protein Transport physiology, Proto-Oncogene Proteins c-mdm2, Tumor Cells, Cultured, Tumor Suppressor Protein p14ARF genetics, Cell Nucleolus metabolism, Nuclear Proteins metabolism, Proto-Oncogene Proteins metabolism, Tumor Suppressor Protein p14ARF metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The ARF tumor suppressor is a nucleolar protein that activates p53-dependent checkpoints by binding Mdm2, a p53 antagonist. Despite persuasive evidence that ARF can bind and inactivate Mdm2 in the nucleoplasm, the prevailing view is that ARF exerts its growth-inhibitory activities from within the nucleolus. We suggest ARF primarily functions outside the nucleolus and provide evidence that it is sequestered and held inactive in that compartment by a nucleolar phosphoprotein, nucleophosmin (NPM). Most cellular ARF is bound to NPM regardless of whether cells are proliferating or growth arrested, indicating that ARF-NPM association does not correlate with growth suppression. Notably, ARF binds NPM through the same domains that mediate nucleolar localization and Mdm2 binding, suggesting that NPM could control ARF localization and compete with Mdm2 for ARF association. Indeed, NPM knockdown markedly enhanced ARF-Mdm2 association and diminished ARF nucleolar localization. Those events correlated with greater ARF-mediated growth suppression and p53 activation. Conversely, NPM overexpression antagonized ARF function while increasing its nucleolar localization. These data suggest that NPM inhibits ARF's p53-dependent activity by targeting it to nucleoli and impairing ARF-Mdm2 association.
- Published
- 2005
- Full Text
- View/download PDF
25. Cytokine-induced phosphoinositide 3-kinase activity promotes Cdk2 activation in factor-dependent hematopoietic cells.
- Author
-
Henry MK, Nimbalkar D, Hohl RJ, and Quelle FW
- Subjects
- Animals, Cell Cycle Proteins metabolism, Cell Division drug effects, Cell Division physiology, Cell Line, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinase 4, Cyclin-Dependent Kinase Inhibitor p27, Cyclin-Dependent Kinases metabolism, Cytokines pharmacology, Enzyme Inhibitors pharmacology, Erythropoietin metabolism, Erythropoietin pharmacology, G1 Phase drug effects, G1 Phase physiology, Hematopoietic Stem Cells drug effects, Interleukin-3 metabolism, Interleukin-3 pharmacology, Mice, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation drug effects, Protein Serine-Threonine Kinases drug effects, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins drug effects, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, S Phase drug effects, S Phase physiology, Signal Transduction drug effects, Signal Transduction physiology, Tumor Suppressor Proteins metabolism, CDC2-CDC28 Kinases metabolism, Cytokines metabolism, Hematopoietic Stem Cells enzymology, Phosphatidylinositol 3-Kinases metabolism
- Abstract
Cytokine growth factors regulate the proliferation of hematopoietic cells through activation of several distinct signaling pathways. We have assessed the contribution of phosphoinositide 3-kinase (PI3K) pathways to erythropoietin (Epo) and interleukin (IL)-3-induced proliferation of factor-dependent hematopoietic cells. Lack of cytokine-induced PI3K activation caused by receptor mutation or treatment with a specific inhibitor (LY294002) did not prevent proliferation but resulted in an increase in the G1 phase content and doubling time of cell cultures. The reduced proliferation of cells lacking cytokine-induced PI3K activity could be partially restored by overexpressing constitutively active Akt. Inhibition of PI3K activity decreased the proportion of cytokine-treated cells entering S phase and was associated with a significant reduction in cytokine-induced phosphorylation and activation of Cdk2. By contrast, Cdk4 activity and p27(Kip1) expression were not significantly altered by inhibition of PI3K. Together, these observations identify a mechanism through which cytokine-activated PI3K contributes to G1 to S phase progression in factor-dependent hematopoietic cells by enhancing the phosphorylation and activation of Cdk2.
- Published
- 2004
- Full Text
- View/download PDF
26. Erythropoietin receptors associate with a ubiquitin ligase, p33RUL, and require its activity for erythropoietin-induced proliferation.
- Author
-
Friedman AD, Nimbalkar D, and Quelle FW
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Cell Division drug effects, Cell Line, Cloning, Molecular, Erythropoietin pharmacology, Humans, In Vitro Techniques, Ligases genetics, Molecular Sequence Data, Mutagenesis, Phosphorylation, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Ligases metabolism, Receptors, Erythropoietin metabolism, Ubiquitin metabolism
- Abstract
The proliferation and survival of hematopoietic cells is strictly regulated by cytokine growth factors that act through receptors of the Type I cytokine receptor family, including erythropoietin (Epo) and its receptor, EpoR. Mitogenic signaling by these receptors depends on activation of Jak tyrosine kinases. However, other required components of this pathway have not been fully identified. In a screen for proteins that interact with EpoR and Jak2, we identified a novel member of the U-box family of ubiquitin ligases. This receptor-associated ubiquitin ligase, RUL, co-precipitated with EpoR from mammalian cells and mediated ubiquitination of EpoR. Also, endogenously expressed RUL was rapidly and transiently phosphorylated on serine after cytokine treatment of factor-dependent hematopoietic cells. Expression of ubiquitin ligase-deficient mutants of RUL inhibited Epo-induced expression of c-myc and bcl-2, two immediate-early genes normally associated with Epo-induced cell growth. Consistent with that finding, expression of mutant RUL also inhibited Epo-dependent proliferation and survival of factor-dependent cells. Together, these observations suggest that RUL is a required component of mitogenic signaling by EpoR. We also show that RUL is phosphorylated in response to growth factors that act through non-cytokine receptors, suggesting that RUL may function as a common regulator of mitogenesis.
- Published
- 2003
- Full Text
- View/download PDF
27. Cytokine activation of phosphoinositide 3-kinase sensitizes hematopoietic cells to cisplatin-induced death.
- Author
-
Nimbalkar D, Henry MK, and Quelle FW
- Subjects
- Animals, Cell Death drug effects, Cell Death physiology, Cell Division drug effects, Cell Line, Cells, Cultured, Chromones pharmacology, Cisplatin administration & dosage, DNA Damage, Drug Synergism, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Erythropoietin administration & dosage, G2 Phase drug effects, Hematopoietic Stem Cells cytology, Interleukin-3 administration & dosage, Mice, Mitosis drug effects, Morpholines pharmacology, Phosphoinositide-3 Kinase Inhibitors, Antineoplastic Agents pharmacology, Cisplatin pharmacology, Erythropoietin pharmacology, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells enzymology, Interleukin-3 pharmacology, Phosphatidylinositol 3-Kinases metabolism
- Abstract
Cytokine growth factors regulate the normal proliferation of hematopoietic cells but can also override irradiation-induced growth arrest checkpoints through activation of a phosphoinositide 3-kinase (PI3K) signaling pathway. In the present study, we assessed the effect that erythropoietin and interleukin-3 have on cisplatin-treated hematopoietic cells. When cultured in the presence of cytokine, cisplatin-treated 32D cells transiently accumulated in a G(2)-M phase arrest and ultimately died by a nonapoptotic mechanism. By comparison, reduction of cytokine-induced PI3K activity, either through cytokine receptor mutation or direct inhibition with LY294002, caused cisplatin-treated cells to enter a biphasic G(1) and G(2)-M arrest. The arrest of these cells coincided with an absence of cyclin-dependent kinase (Cdk)1 and Cdk2 activity and significantly reduced cell death during cisplatin treatment. Indeed, LY294002 treatment during cisplatin exposure allowed the recovery of a viable, proliferating cell population after removal of cisplatin. In contrast, Cdks remained active in the G(2)-M-arrested population of cisplatin-treated cells with continuous cytokine activation of PI3K, and even transient exposure to cisplatin resulted in death of the entire population. These data suggest that cytokine activation of PI3K signaling pathways overrides cisplatin-induced growth arrest checkpoints, thereby sensitizing hematopoietic cells to DNA damage-induced death.
- Published
- 2003
28. Dna damage-induced G(1) arrest in hematopoietic cells is overridden following phosphatidylinositol 3-kinase-dependent activation of cyclin-dependent kinase 2.
- Author
-
Eapen AK, Henry MK, Quelle DE, and Quelle FW
- Subjects
- Animals, Cell Line, Cyclin-Dependent Kinase 2, DNA Damage, Enzyme Activation, Hematopoiesis physiology, Mice, Signal Transduction, CDC2-CDC28 Kinases, Cyclin-Dependent Kinases physiology, G1 Phase physiology, Phosphatidylinositol 3-Kinases physiology, Protein Serine-Threonine Kinases physiology
- Abstract
Exposure of hematopoietic cells to DNA-damaging agents induces p53-independent cell cycle arrest at a G(1) checkpoint. Previously, we have shown that this growth arrest can be overridden by cytokine growth factors, such as erythropoietin or interleukin-3, through activation of a phosphatidylinositol 3-kinase (PI 3-kinase)/Akt-dependent signaling pathway. Here, we show that gamma-irradiated murine myeloid 32D cells arrest in G(1) with active cyclin D-cyclin-dependent kinase 4 (Cdk4) but with inactive cyclin E-Cdk2 kinases. The arrest was associated with elevated levels of the Cdk inhibitors p21(Cip1) and p27(Kip1), yet neither was associated with Cdk2. Instead, irradiation-induced inhibition of cyclin E-Cdk2 correlated with absence of the activating threonine-160 phosphorylation on Cdk2. Cytokine treatment of irradiated cells induced Cdk2 phosphorylation and activation, and cells entered into S phase despite sustained high-level expression of p21 and p27. Notably, the PI 3-kinase inhibitor, LY294002, completely blocked cytokine-induced Cdk2 activation and cell growth in irradiated 32D cells but not in nonirradiated cells. Together, these findings demonstrate a novel mechanism underlying the DNA damage-induced G(1) arrest of hematopoietic cells, that is, inhibition of Cdk2 phosphorylation and activation. These observations link PI 3-kinase signaling pathways with the regulation of Cdk2 activity.
- Published
- 2001
- Full Text
- View/download PDF
29. DNA damage-induced cell-cycle arrest of hematopoietic cells is overridden by activation of the PI-3 kinase/Akt signaling pathway.
- Author
-
Henry MK, Lynch JT, Eapen AK, and Quelle FW
- Subjects
- Animals, Cell Cycle radiation effects, Cell Line, Enzyme Activation, Erythropoietin pharmacology, Gamma Rays, Hematopoietic Stem Cells radiation effects, Interleukin-3 pharmacology, Mice, Mutation, Phosphatidylinositol 3-Kinases metabolism, Phosphatidylinositol 3-Kinases pharmacology, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins pharmacology, Proto-Oncogene Proteins c-akt, Receptors, Erythropoietin genetics, Signal Transduction drug effects, Cell Cycle drug effects, DNA Damage physiology, Hematopoietic Stem Cells physiology, Phosphatidylinositol 3-Kinases physiology, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins physiology
- Abstract
Exposure of hematopoietic cells to DNA-damaging agents induces cell-cycle arrest at G1 and G2/M checkpoints. Previously, it was shown that DNA damage-induced growth arrest of hematopoietic cells can be overridden by treatment with cytokine growth factors, such as erythropoietin (EPO) or interleukin-3 (IL-3). Here, the cytokine-activated signaling pathways required to override G1 and G2/M checkpoints induced by gamma-irradiation (gamma-IR) are characterized. Using factor-dependent myeloid cells stably expressing EPO receptor (EPO-R) mutants, it is shown that removal of a minimal domain required for PI-3K signaling abrogated the ability of EPO to override gamma-IR-induced cell-cycle arrest. Similarly, the ability of cytokines to override gamma-IR-induced arrest was abolished by an inhibitor of PI-3K (LY294002) or by overexpression of dominant-negative Akt. Moreover, the ability of EPO to override these checkpoints in cells expressing defective EPO-R mutants could be restored by overexpression of a constitutively active Akt. Thus, activation of a PI-3K/Akt signaling pathway is required for cytokine-dependent suppression of DNA-damage induced checkpoints. Together, these findings suggest a novel role for PI-3K/Akt pathways in the modulation of growth arrest responses to DNA damage in hematopoietic cells. (Blood. 2001;98:834-841)
- Published
- 2001
- Full Text
- View/download PDF
30. Cytokine rescue of p53-dependent apoptosis and cell cycle arrest is mediated by distinct Jak kinase signaling pathways.
- Author
-
Quelle FW, Wang J, Feng J, Wang D, Cleveland JL, Ihle JN, and Zambetti GP
- Subjects
- Animals, Cell Division, Cell Line, DNA Damage, Enzyme Activation, Erythropoietin metabolism, Humans, Interleukin-3 pharmacology, Janus Kinase 2, Mice, Proto-Oncogene Proteins c-bcl-2 metabolism, Receptors, Erythropoietin metabolism, Serum Albumin, Bovine pharmacology, bcl-X Protein, Apoptosis, Cell Cycle, Interleukin-3 metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Signal Transduction, Tumor Suppressor Protein p53 metabolism
- Abstract
Exposure of hematopoietic progenitors to gamma-irradiation (IR) induces p53-dependent apoptosis and a p53-independent G2/M cell cycle arrest. These responses to DNA-damage can be inhibited by treatment with cytokine growth factors. Here we report that gamma-IR-induced apoptosis and cell cycle arrest are suppressed by specific cytokines (e.g., erythropoietin and interleukin-3) and that activation of the Jak kinase is necessary and sufficient for these effects. Using myleoid cells expressing a series of erythropoietin receptor (EpoR) mutants, we have demonstrated that Jak kinase-dependent signals initiated from the membrane proximal domain of EpoR were sufficient to prevent IR-induced apoptotic cell death, but failed to prevent cell cycle arrest. Cell survival by Epo did not require activation of other known signaling pathways including PI-3 kinase, PLC-gamma, Ras or Stats. Signaling targets of Jak kinase pathways included members of the Bcl-2 family of anti-apoptotic proteins, and enforced expression of Bcl-2 or Bcl-xL was as effective as cytokine treatment in blocking IR-induced apoptosis but did not prevent growth arrest. A distinct signal derived from a membrane distal domain of EpoR is required to overcome growth arrest associated with DNA damage. These findings functionally link the Jak signaling pathway to suppression of p53-mediated cell death by cytokines and demonstrate that the apoptotic and growth arrest responses to DNA damage in hematopoietic cells are modulated by distinct, cytokine specific signal transduction pathways.
- Published
- 1998
- Full Text
- View/download PDF
31. Jaks and Stats in cytokine signaling.
- Author
-
Ihle JN, Nosaka T, Thierfelder W, Quelle FW, and Shimoda K
- Subjects
- Animals, JNK Mitogen-Activated Protein Kinases, Mice, STAT4 Transcription Factor, STAT5 Transcription Factor, STAT6 Transcription Factor, Calcium-Calmodulin-Dependent Protein Kinases physiology, Cytokines physiology, DNA-Binding Proteins physiology, Milk Proteins, Mitogen-Activated Protein Kinases, Signal Transduction physiology, Trans-Activators physiology
- Abstract
Hematopoiesis is regulated through the binding of cytokines to receptors of the cytokine receptor superfamily. Although lacking catalytic domains, members of the cytokine receptor superfamily mediate ligand-dependent activation of protein tyrosine phosphorylation through their association and activation of members of the Janus kinase (Jak) family of protein tyrosine kinases. The activated Jaks phosphorylate the receptors which creates docking sites for SH2-containing signaling proteins which are tyrosine phosphorylated following their association with the complex. Among the substrates of tyrosine phosphorylation are members of the signal transducers and activators of the transcription family of proteins (Stats). Various cytokines induce the tyrosine phosphorylation and activation of one or more of the seven family members. The pattern of Stat activation provides a level of cytokine individuality that is not observed in the activation of other signaling pathways. The role of various Stats in the biological responses to cytokines has been assessed through the analysis of receptor mutations which disrupt Stat activation and more recently by disruption of the genes in mice. Our results have demonstrated that the activation of Stat5a and Stat5b by erythropoietin is critical for the activation of a number of immediate early genes but is not required for a mitogenic response. Mice in which the genes for Stat4 and Stat6 are disrupted are viable but lack functions that are mediated by interleukin 12 (IL-12) or IL-4, respectively, suggesting that these Stats perform very specific functions in immune responses.
- Published
- 1997
- Full Text
- View/download PDF
32. Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene.
- Author
-
Shimoda K, van Deursen J, Sangster MY, Sarawar SR, Carson RT, Tripp RA, Chu C, Quelle FW, Nosaka T, Vignali DA, Doherty PC, Grosveld G, Paul WE, and Ihle JN
- Subjects
- Animals, Base Sequence, Cell Differentiation genetics, Cell Differentiation physiology, Cell Line, Cells, Cultured, DNA Probes, Gene Targeting, Immunity genetics, Immunity physiology, Lymphoid Tissue immunology, Mice, Molecular Sequence Data, STAT6 Transcription Factor, Th2 Cells immunology, Trans-Activators genetics, Immunoglobulin Class Switching, Immunoglobulin E immunology, Interleukin-4 immunology, Th2 Cells cytology, Trans-Activators physiology
- Abstract
Signal transducers and activators of transcription (Stats) are activated by tyrosine phosphorylation in response to cytokines, and are thought to mediate many of their functional responses. Stat6 is activated in response to interleukin (IL)-4 and may contribute to various functions including mitogenesis, T-helper cell differentiation and immunoglobulin isotype switching. To evaluate the role of Stat6, we generated Stat6-null mice (Stat6 -/-) by gene disruption in embryonic stem cells. The mice were viable, indicating the lack of a non-redundant function in normal development. Although naive lymphoid cell development was normal, Stat6 -/- mice were deficient in IL-4-mediated functions including Th2 helper T-cell differentiation, expression of cell surface markers, and immunoglobulin class switching to IgE. In contrast, IL-4-mediated proliferation was only partly affected.
- Published
- 1996
- Full Text
- View/download PDF
33. Erythropoietin induces activation of Stat5 through association with specific tyrosines on the receptor that are not required for a mitogenic response.
- Author
-
Quelle FW, Wang D, Nosaka T, Thierfelder WE, Stravopodis D, Weinstein Y, and Ihle JN
- Subjects
- Amino Acid Sequence, Animals, Cell Division drug effects, Cell Line, DNA-Binding Proteins genetics, Enzyme Activation, Humans, Janus Kinase 2, Molecular Sequence Data, Mutation, Protein-Tyrosine Kinases genetics, Protein-Tyrosine Kinases metabolism, Receptors, Antigen, T-Cell, gamma-delta drug effects, Receptors, Antigen, T-Cell, gamma-delta genetics, Receptors, Antigen, T-Cell, gamma-delta metabolism, Receptors, Erythropoietin drug effects, Receptors, Erythropoietin genetics, STAT5 Transcription Factor, Trans-Activators genetics, Tumor Suppressor Proteins, Cell Division genetics, DNA-Binding Proteins metabolism, Erythropoietin pharmacology, Milk Proteins, Proto-Oncogene Proteins, Receptors, Erythropoietin metabolism, Trans-Activators metabolism, Tyrosine metabolism
- Abstract
The cytoplasmic domain of the erythropoietin receptor (EpoR) contains a membrane-distal region that is dispensable for mitogenesis but is required for the recruitment and tyrosine phosphorylation of a variety of signaling proteins. The membrane-proximal region of 96 amino acids is necessary and sufficient for mitogenesis as well as Jak2 activation, induction of c-fos, c-myc, cis, the T-cell receptor gamma locus (TCR-gamma), and c-pim-1. The studies presented here demonstrate that this region is also necessary and sufficient for the activation of Stat5A and Stat5B. The membrane-proximal domain contains a single tyrosine, Y-343, which when mutated eliminates the ability of the receptor to couple Epo binding to the activation of Stat5. Furthermore, peptide competitions demonstrate that this site, when phosphorylated, can disrupt Stat5 DNA binding activity, consistent with a role of Y-343 as a site of recruitment to the receptor. Cells expressing the truncated, Y343F mutant (a mutant with a Y-to-F alteration at position 343) proliferate in response to Epo in a manner comparable to that of the controls. However, in these cells, Epo stimulation does not induce the appearance of transcripts for cis, TCR-gamma, or c-fos, suggesting a role for Stat5 in their regulation.
- Published
- 1996
- Full Text
- View/download PDF
34. Interleukin-9 induces tyrosine phosphorylation of insulin receptor substrate-1 via JAK tyrosine kinases.
- Author
-
Yin T, Keller SR, Quelle FW, Witthuhn BA, Tsang ML, Lienhard GE, Ihle JN, and Yang YC
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Insulin Receptor Substrate Proteins, Interleukin-4 pharmacology, Janus Kinase 1, Janus Kinase 2, Janus Kinase 3, Kinetics, Mice, Phosphoproteins isolation & purification, Phosphorylation, Phosphotyrosine, Proteins metabolism, Recombinant Proteins metabolism, T-Lymphocytes, TYK2 Kinase, Transfection, Tyrosine analogs & derivatives, Tyrosine analysis, Interleukin-9 pharmacology, Phosphoproteins metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins
- Abstract
Interleukin (IL)-9 stimulates the proliferation of a variety of hematopoietic lineages through its interaction with a receptor of the cytokine receptor superfamily. In the studies presented here, we have begun to characterize the downstream signaling pathways activated by IL-9. In addition to the activation of JAK1 and JAK3 tyrosine kinases, IL-9, unlike most hematopoietic cytokines but similar to IL-4, induces the tyrosine phosphorylation of a 170-kDa protein that is related to the insulin receptor substrate-1 (IRS-1). We further demonstrate for the first time that IRS-1 is not only associated with JAK1 but also tyrosine phosphorylated and functionally involved in IL-9 signaling in TS1 lymphocytes transfected with the murine IRS-1 cDNA. Cotransfection studies and in vitro experiments directly demonstrate that JAK1, JAK2, or JAK3 is capable of tyrosine phosphorylating IRS-1, suggesting a functional role for these kinases in vivo. Lastly, we demonstrate that IL-9 induces the tyrosine phosphorylation of Stat3 and in this regard differs from IL-4, which triggers tyrosine phosphorylation of Stat6. Taken together, these results strongly suggest that IL-9 and IL-4 utilize common and unique signaling pathways via inducing the similar and distinct tyrosine-phosphorylated proteins.
- Published
- 1995
- Full Text
- View/download PDF
35. Phosphorylation and activation of the DNA binding activity of purified Stat1 by the Janus protein-tyrosine kinases and the epidermal growth factor receptor.
- Author
-
Quelle FW, Thierfelder W, Witthuhn BA, Tang B, Cohen S, and Ihle JN
- Subjects
- Amino Acid Sequence, Animals, Blotting, Western, Cell Line, DNA-Binding Proteins biosynthesis, DNA-Binding Proteins isolation & purification, Humans, Janus Kinase 1, Janus Kinase 2, Mammals, Mice, Molecular Sequence Data, Peptide Mapping, Phosphopeptides chemistry, Phosphopeptides isolation & purification, Phosphorylation, Protein Biosynthesis, Protein-Tyrosine Kinases biosynthesis, Protein-Tyrosine Kinases isolation & purification, Proteins isolation & purification, Proteins metabolism, Recombinant Proteins biosynthesis, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, STAT1 Transcription Factor, Sequence Homology, Amino Acid, Sheep, Spodoptera, TYK2 Kinase, Trans-Activators biosynthesis, Trans-Activators isolation & purification, Transfection, Tyrosine, DNA-Binding Proteins metabolism, ErbB Receptors metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Trans-Activators metabolism
- Abstract
The activation of Janus protein-tyrosine kinases (Jaks) and the subsequent phosphorylation and activation of latent signal transducers and activators of transcription (Stats) are common elements in signal transduction through the cytokine receptor superfamily. To assess the role and specificity of Jaks in Stat activation, we have utilized baculovirus expression systems to produce Stat1 and the Jaks. Co-expression of Stat1 with Tyk2, Jak1, or Jak2 resulted in the specific tyrosine phosphorylation of Stat1 at Tyr701, the residue phosphorylated in mammalian cells stimulated with interferon gamma. Alternatively, Stat1, purified to apparent homogeneity from insect cell extracts, was phosphorylated at Tyr701 in Jak immune complex kinase reactions. Phosphorylation of purified Stat1 was necessary and sufficient for the acquisition of DNA binding activity. The specificity in both systems was indicated by the inability of a Jak2 catalytically inactive mutant (Jak2-Glu882) or the Tec protein-tyrosine kinase to phosphorylate Stat1. However, immune complex-purified epidermal growth factor receptor was capable of phosphorylating purified Stat1 at Tyr701 and activating its DNA binding activity in in vitro reactions.
- Published
- 1995
- Full Text
- View/download PDF
36. Distribution of the mammalian Stat gene family in mouse chromosomes.
- Author
-
Copeland NG, Gilbert DJ, Schindler C, Zhong Z, Wen Z, Darnell JE Jr, Mui AL, Miyajima A, Quelle FW, and Ihle JN
- Subjects
- Animals, Crosses, Genetic, Female, Male, Mammals genetics, Mice, Oligonucleotide Probes, Restriction Mapping, Transcription Factors biosynthesis, Chromosome Mapping, Mice, Inbred C57BL genetics, Multigene Family, Muridae genetics, Transcription Factors genetics
- Abstract
Studies of transcriptional activation by interferons and a variety of cytokines have led to the identification of a family of proteins that serve as signal transducers and activators of transcription, Stats. Here, we report that the seven mouse Stat loci map in three clusters, with each cluster located on a different mouse autosome. The data suggest that the family has arisen via a tandem duplication of the ancestral locus, followed by dispersion of the linked loci to different mouse chromosomes.
- Published
- 1995
- Full Text
- View/download PDF
37. Cloning of murine Stat6 and human Stat6, Stat proteins that are tyrosine phosphorylated in responses to IL-4 and IL-3 but are not required for mitogenesis.
- Author
-
Quelle FW, Shimoda K, Thierfelder W, Fischer C, Kim A, Ruben SM, Cleveland JL, Pierce JH, Keegan AD, and Nelms K
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Line, Cloning, Molecular, Humans, Mice, Mitosis drug effects, Molecular Sequence Data, Phosphorylation, STAT6 Transcription Factor, Sequence Analysis, Trans-Activators metabolism, Transcription Factors isolation & purification, Transcription Factors metabolism, Interleukin-3 pharmacology, Interleukin-4 pharmacology, Trans-Activators genetics, Transcription Factors genetics
- Abstract
By searching a database of expressed sequences, we identified a member of the signal transducers and activators of transcription (Stat) family of proteins. Human and murine full-length cDNA clones were obtained and sequenced. The sequence of the human cDNA was identical to the recently published sequence for interleukin-4 (IL-4)-Stat (J. Hou, U. Schindler, W.J. Henzel, T.C. Ho, M. Brasseur, and S. L. McKnight, Science 265:1701-1706, 1994), while the murine Stat6 amino acid and nucleotide sequences were 83 and 84% identical to the human sequences, respectively. Using Stat6-specific antiserum, we demonstrated that Stat6 is rapidly tyrosine phosphorylated following stimulation of appropriate cell lines with IL-4 or IL-3 but is not detectably phosphorylated following stimulation with IL-2, IL-12, or erythropoietin. In contrast, IL-2, IL-3, and erythropoietin induce the tyrosine phosphorylation of Stat5 while IL-12 uniquely induces the tyrosine phosphorylation of Stat4. Inducible tyrosine phosphorylation of Stat6 requires the membrane-distal region of the IL-4 receptor alpha chain. This region of the receptor is not required for cell growth, demonstrating that Stat6 tyrosine phosphorylation does not contribute to mitogenesis.
- Published
- 1995
- Full Text
- View/download PDF
38. Signaling through the hematopoietic cytokine receptors.
- Author
-
Ihle JN, Witthuhn BA, Quelle FW, Yamamoto K, and Silvennoinen O
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, DNA genetics, DNA metabolism, Humans, Molecular Sequence Data, Molecular Structure, Protein-Tyrosine Kinases chemistry, Protein-Tyrosine Kinases genetics, Protein-Tyrosine Kinases physiology, Signal Transduction physiology, Trans-Activators genetics, Trans-Activators physiology, Hematopoiesis physiology, Receptors, Cytokine physiology
- Abstract
Hematopoiesis is regulated through the interaction of a variety of growth factors with specific receptors of the cytokine receptor superfamily. Although lacking catalytic domains, all the receptors couple ligand binding to the rapid induction of protein tyrosine phosphorylation. This is mediated through a novel family of protein tyrosine kinases termed the Janus kinases (Jaks) which associate with the receptors and are activated following ligand binding. Depending upon the cytokine/receptor system, one or more of the four known Jaks (Jak1, Jak2, Jak3, Tyk2) is/are involved. The activated Jaks phosphorylate both themselves and the receptor subunits, creating docking sites for SH2-containing proteins including SHC, which couples receptor engagement to activation of the ras pathway, and HCP, a protein tyrosine phosphatase which negatively affects the response. In addition, the Jaks phosphorylate one or more of a family of signal transducers and activators of transcription (Stats). Phosphorylation of Stats induces their nuclear translocation and DNA-binding activity. Activation of Stats is independent of activation of the ras pathway and represents a novel signaling pathway correlated with mitogenesis.
- Published
- 1995
- Full Text
- View/download PDF
39. JAK2 associates with the beta c chain of the receptor for granulocyte-macrophage colony-stimulating factor, and its activation requires the membrane-proximal region.
- Author
-
Quelle FW, Sato N, Witthuhn BA, Inhorn RC, Eder M, Miyajima A, Griffin JD, and Ihle JN
- Subjects
- Animals, Cell Line, DNA Mutational Analysis, Enzyme Activation, Humans, Janus Kinase 2, Macromolecular Substances, Moths, Mutagenesis, Phosphorylation, Protein-Tyrosine Kinases biosynthesis, Protein-Tyrosine Kinases isolation & purification, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor biosynthesis, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor isolation & purification, Signal Transduction, Transfection, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
The high-affinity receptor for granulocyte-macrophage colony-stimulating factor (GM-CSF) consists of a unique alpha chain and a beta c subunit that is shared with the receptors for interleukin-3 (IL-3) and IL-5. Two regions of the beta c chain have been defined; these include a membrane-proximal region of the cytoplasmic domain that is required for mitogenesis and a membrane-distal region that is required for activation of Ras, Raf-1, mitogen-activated protein kinase, and S6 kinase. Recent studies have implicated the cytoplasmic protein tyrosine kinase JAK2 in signalling through a number of the cytokine receptors, including the IL-3 and erythropoietin receptors. In the studies described here, we demonstrate that GM-CSF stimulation of cells induces the tyrosine phosphorylation of JAK2 and activates its in vitro kinase activity. Mutational analysis of the beta c chain demonstrates that only the membrane-proximal 62 amino acids of the cytosolic domain are required for JAK2 activation. Thus, JAK2 activation is correlated with induction of mitogenesis but does not, alone, activate the Ras pathway. Carboxyl truncations of the alpha chain, which inactivate the receptor for mitogenesis, are unable to mediate GM-CSF-induced JAK2 activation. Using baculovirus-expressed proteins, we further demonstrate that JAK2 physically associates with the beta c chain but not with the alpha chain. Together, the results further support the hypothesis that the JAK family of kinase are critical to coupling cytokine binding to tyrosine phosphorylation and ultimately mitogenesis.
- Published
- 1994
- Full Text
- View/download PDF
40. Stat4, a novel gamma interferon activation site-binding protein expressed in early myeloid differentiation.
- Author
-
Yamamoto K, Quelle FW, Thierfelder WE, Kreider BL, Gilbert DJ, Jenkins NA, Copeland NG, Silvennoinen O, and Ihle JN
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Differentiation, Cell Line, Chlorocebus aethiops, Conserved Sequence, Crosses, Genetic, DNA Primers, DNA-Binding Proteins genetics, DNA-Binding Proteins isolation & purification, Female, Hematopoietic Stem Cells cytology, Male, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Muridae, Oligonucleotide Probes, Open Reading Frames, Organ Specificity, Polymerase Chain Reaction, STAT4 Transcription Factor, Sequence Homology, Amino Acid, Trans-Activators genetics, Trans-Activators isolation & purification, Transfection, Chromosome Mapping, DNA-Binding Proteins biosynthesis, Hematopoietic Stem Cells metabolism, Multigene Family, Trans-Activators biosynthesis
- Abstract
Interferon regulation of gene expression is dependent on the tyrosine phosphorylation and activation of the DNA-binding activity of two related proteins of 91 kDa (STAT1) and/or 113 kDa (STAT2). Recent studies have suggested that these proteins are substrates of Janus kinases and that proteins related in STAT1 are involved in a number of signalling pathways, including those activated in myeloid cells by erythropoietin and interleukin-3 (IL-3). To clone STAT-related proteins from myeloid cells, degenerate oligonucleotides were used in PCRs to identify novel family members expressed in myeloid cells. This approach allowed the identification and cloning of the Stat4 gene, which is 52% identical to STAT1. Unlike STAT1, Stat4 expression is restricted but includes myeloid cells and spermatogonia. In the erythroid lineage, Stat4 expression is differentially regulated during differentiation. Functionally, Stat4 has the properties of other STAT family genes. In particular, cotransfection of expression constructs for Stat4 and Jak1 and Jak2 results in the tyrosine phosphorylation of Stat4 and the acquisition of the ability to bind to the gamma interferon (IFN-gamma)-activated sequence of the interferon regulatory factor 1 (IRF-1) gene. Stat4 is located on mouse chromosome 1 and is tightly linked to the Stat1 gene, suggesting that the genes arose by gene duplication. Unlike Stat1, neither IFN-alpha nor IFN-gamma activates Stat4. Nor is Stat4 activated in myeloid cells by a number of cytokines, including erythropoietin, IL-3, granulocyte colony-stimulating factor, stem cell factor, colon-stimulating factor 1, hepatocyte growth factor, IL-2, IL-4, and IL-6.
- Published
- 1994
- Full Text
- View/download PDF
41. Signaling by the cytokine receptor superfamily: JAKs and STATs.
- Author
-
Ihle JN, Witthuhn BA, Quelle FW, Yamamoto K, Thierfelder WE, Kreider B, and Silvennoinen O
- Subjects
- Amino Acid Sequence, Cytokines physiology, Janus Kinase 1, Janus Kinase 2, Molecular Sequence Data, Protein-Tyrosine Kinases chemistry, Sequence Homology, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins, Receptors, Cytokine physiology, Signal Transduction, Transcription Factors physiology
- Abstract
A variety of cytokines, lymphokines and growth factors function by interacting with receptors that are members of the cytokine receptor superfamily. These receptors share extracellular motifs and have limited similarity in their cytoplasmic domains. Although lacking catalytic domains, this family of receptors couples ligand binding with the induction of tyrosine phosphorylation. Recent studies have shown that this is mediated by members of the Janus kinase (JAK) family of cytoplasmic protein tyrosine kinases. JAKs physically associate with the membrane-proximal region of the ligand-bound receptor, leading to their tyrosine phosphorylation and activation. The activated JAKs phosphorylate the receptors as well as cytoplasmic proteins belonging to a family of transcription factors called the signal transducers and activators of transcription (STATs), providing a novel signaling pathway that is shared by all members of the cytokine receptor superfamily.
- Published
- 1994
- Full Text
- View/download PDF
42. Cytokine receptors and signal transduction.
- Author
-
Ihle JN, Witthuhn B, Tang B, Yi T, and Quelle FW
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Humans, Molecular Sequence Data, Phosphotyrosine, Protein-Tyrosine Kinases physiology, Receptors, Colony-Stimulating Factor chemistry, Receptors, Colony-Stimulating Factor physiology, Receptors, Cytokine chemistry, Receptors, Cytokine genetics, Tyrosine analogs & derivatives, Receptors, Cytokine physiology, Signal Transduction
- Abstract
The past few years have seen an explosion in the identification, cloning and characterization of cytokines and their receptors. The pleiotropic effects of many of the growth factors and the considerable redundancy in the actions of growth factors have contributed to a mass of descriptive literature that often seems to defy summary. Only recently have common concepts begun to emerge. First, cytokines mediate their effects through a large family of receptors that have evolved from a common progenitor and retain structural and functional similarities. Within the haematopoietic system, the cytokines are not usually instructive in differentiation, but rather supportive, and may contribute to some differentiation-specific responses. The patterns of expression of cytokine receptors are therefore a product of differentiation and provide for changes in physiological regulation. The second important concept that is emerging is that the cytokines mediate their mitogenic effects through a common signal-transducing pathway involving tyrosine phosphorylation. Thus, although the cytokine receptor superfamily members do not have intrinsic protein tyrosine kinase activity, by coupling to activation of tyrosine phosphorylation they may affect cell growth by pathways that are common with the large family of growth factor receptors that contain intrinsic protein tyrosine kinase activity. The coupling of cytokine binding to tyrosine phosphorylation and mitogenesis requires a relatively small membrane-proximal domain of the receptors. This region has limited sequence similarity which may be required for the association of individual receptors with an appropriate kinase. Activation of kinase activity results from the dimerization or oligomerization of receptor homodimers or heterodimers. Again this requirement is similar to that seen with the growth factor receptors which have intrinsic protein tyrosine kinase activity. The protein tyrosine kinases that couple cytokine binding to tyrosine phosphorylation are members of the Jak family of kinases. The ubiquitous expression of these kinases provides a common cellular background on which the cytokine receptors can function and on which unique functionally distinct receptors have evolved. In particular, tyk2 is required for the responses initiated by IFN-alpha while Jak2 has been implicated in the responses to G-CSF, IL-3, EPO, growth hormone, prolactin and IFN-gamma.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
43. Association and activation of Jak-Tyk kinases by CNTF-LIF-OSM-IL-6 beta receptor components.
- Author
-
Stahl N, Boulton TG, Farruggella T, Ip NY, Davis S, Witthuhn BA, Quelle FW, Silvennoinen O, Barbieri G, and Pellegrini S
- Subjects
- Animals, Cell Line, Ciliary Neurotrophic Factor, Cytokine Receptor gp130, Cytokines metabolism, Enzyme Activation, Janus Kinase 1, Janus Kinase 2, Leukemia Inhibitory Factor, Nerve Tissue Proteins metabolism, Nerve Tissue Proteins pharmacology, Oncostatin M, Peptides metabolism, Peptides pharmacology, Phosphorylation, Proteins metabolism, Receptor, Ciliary Neurotrophic Factor, Receptors, Growth Factor metabolism, Receptors, Interleukin metabolism, Receptors, Interleukin-6, Receptors, OSM-LIF, Receptors, Oncostatin M, Tyrosine metabolism, Antigens, CD, Cytokines pharmacology, Growth Inhibitors, Interleukin-6, Lymphokines, Membrane Glycoproteins metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Receptors, Cytokine metabolism
- Abstract
A recently defined family of cytokines, consisting of ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), oncostatin M (OSM), and interleukin-6 (IL-6), utilize the Jak-Tyk family of cytoplasmic tyrosine kinases. The beta receptor components for this cytokine family, gp130 and LIF receptor beta, constitutively associate with Jak-Tyk kinases. Activation of these kinases occurs as a result of ligand-induced dimerization of the receptor beta components. Unlike other cytokine receptors studied to date, the receptors for the CNTF cytokine family utilize all known members of the Jak-Tyk family, but induce distinct patterns of Jak-Tyk phosphorylation in different cell lines.
- Published
- 1994
- Full Text
- View/download PDF
44. Protein tyrosine phosphorylation in the regulation of hematopoiesis by receptors of the cytokine-receptor superfamily.
- Author
-
Ihle JN, Witthuhn BA, Quelle FW, Silvennoinen O, Tang B, and Yi T
- Subjects
- Animals, Humans, Phosphorylation, Protein Structure, Tertiary, Receptors, Erythropoietin chemistry, Signal Transduction physiology, Structure-Activity Relationship, Hematopoiesis physiology, Protein-Tyrosine Kinases metabolism, Receptors, Cytokine physiology
- Abstract
Cytokines regulate the growth and differentiation of hematopoietic cells through their interaction with receptors of the cytokine receptor superfamily. This family of receptors has conserved motifs in the extracellular domain but share only limited similarity in the cytoplasmic domains. Although lacking catalytic domains, a variety of studies demonstrate that the cytokine receptors function by coupling ligand binding to induction of tyrosine phosphorylation. Recent studies have shown that the JAK family of kinases associate with cytokine receptors and are activated by ligand binding. Interaction occurs with the membrane proximal region of the cytoplasmic domain, a region that has been found to be essential for mitogenesis. One of the substrates of tyrosine phosphorylation is the receptor and, in the case of the receptor for Epo, the membrane distal region of the cytoplasmic domain is phosphorylated. Once phosphorylated, this site becomes a binding site for the amino-terminal SH2 domain of hematopoietic cell phosphatase (HCP). HCP is an important negative regulator of hematopoietic cell growth and its recruitment to the receptor complex is speculated to be important for this effect. The role of HCP is best indicated by the observation that the murine mutation, motheaten, is due to a mutation that results in the inability to make HCP. Motheaten mice die soon after birth due to the overproliferation of a variety of hematopoietic lineages. Together the results demonstrate an essential role in both protein tyrosine phosphorylation and de-phosphorylation in the growth regulation of hematopoiesis.
- Published
- 1994
45. Complementation by the protein tyrosine kinase JAK2 of a mutant cell line defective in the interferon-gamma signal transduction pathway.
- Author
-
Watling D, Guschin D, Müller M, Silvennoinen O, Witthuhn BA, Quelle FW, Rogers NC, Schindler C, Stark GR, and Ihle JN
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte metabolism, CD2 Antigens, Cell Line, DNA-Binding Proteins metabolism, Enzyme Activation, Flow Cytometry, Genetic Complementation Test, Humans, Interferon-Stimulated Gene Factor 3, Interferon-Stimulated Gene Factor 3, gamma Subunit, Interferon-alpha metabolism, Interferon-beta metabolism, Interferon-gamma genetics, Janus Kinase 2, L Cells, Mice, Mutation, Phosphorylation, Precipitin Tests, Protein-Tyrosine Kinases genetics, RNA, Messenger biosynthesis, Receptors, Immunologic metabolism, Transcription Factors metabolism, Transfection, Tyrosine metabolism, Interferon-gamma metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Signal Transduction genetics
- Abstract
Interferons (IFNs) alpha/beta (type I) and gamma (type II) bind to distinct cell surface receptors, inducing transcription of overlapping sets of genes by intracellular pathways that have recently attracted much attention. Previous studies using cell lines selected for their inability to respond to IFN-alpha (ref. 4) have shown that the protein kinase Tyk2 plays a central role in the IFN alpha/beta response. Here we report the isolation of the cell line gamma 1A, selected for its inability to express IFN-gamma-inducible cell-surface markers, that is deficient in all aspects of the IFN-gamma response tested, but responds normally to IFNs alpha and beta. The mutant cells can be complemented by the expression of another member of the JAK family of protein tyrosine kinases, JAK2 (refs 6-9). Unlike IFNs alpha and beta, IFN-gamma induces rapid tyrosine phosphorylation of JAK2 in wild-type cells, and JAK2 immunoprecipitates from these cells show tyrosine kinase activity. These responses are absent in gamma 1A cells. JAK2 is therefore required for the response to IFN-gamma but not to IFNs alpha and beta.
- Published
- 1993
- Full Text
- View/download PDF
46. Signal transduction through the receptor for erythropoietin.
- Author
-
Ihle JN, Quelle FW, and Miura O
- Subjects
- Amino Acid Sequence, Animals, Humans, Molecular Sequence Data, Protein-Tyrosine Kinases physiology, Receptors, Erythropoietin chemistry, Receptors, Erythropoietin physiology, Signal Transduction
- Abstract
Erythropoietin (EPO) supports the proliferation and differentiation of erythroid lineage cells. The receptor for erythropoietin is a member of the cytokine receptor family. Introduction of EPOR into IL-3 dependent cells confers the ability to proliferate in response to EPO. Associated with this, EPO induces the expression of a number of immediate-early response genes. Mutagenesis studies have addressed the function role of various motifs and domains in receptor function and established essential roles for the conserved cysteines and the WSXWS motif. The signal transducing pathways activated by EPOR include induction of tyrosine phosphorylation. Within the cytoplasmic domain a relatively small membrane proximal region is essential for induction of tyrosine phosphorylation, expression of immediate early genes and for mitogenesis. The role of various kinases in this response is discussed as well as an assessment of potential substrates of tyrosine phosphorylation.
- Published
- 1993
- Full Text
- View/download PDF
47. Structure of the murine Jak2 protein-tyrosine kinase and its role in interleukin 3 signal transduction.
- Author
-
Silvennoinen O, Witthuhn BA, Quelle FW, Cleveland JL, Yi T, and Ihle JN
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Bone Marrow enzymology, Cell Line, Cells, Cultured, Cloning, Molecular, DNA genetics, DNA metabolism, Janus Kinase 2, Mice, Molecular Sequence Data, Monocytes enzymology, Oligodeoxyribonucleotides, Polymerase Chain Reaction methods, Protein Biosynthesis, Protein-Tyrosine Kinases genetics, Protein-Tyrosine Kinases isolation & purification, RNA, Messenger isolation & purification, RNA, Messenger metabolism, Signal Transduction drug effects, Transcription, Genetic, Interleukin-3 pharmacology, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Signal Transduction physiology
- Abstract
Interleukin 3 (IL-3) regulates the proliferation and differentiation of hematopoietic cells. Although the IL-3 receptor chains lack kinase catalytic domains, IL-3 induces tyrosine phosphorylation of cellular proteins. To investigate the potential role of the JAK family of protein-tyrosine kinases in IL-3 signal transduction, we have obtained full-length cDNA clones for murine Jak1 and Jak2 protein-tyrosine kinases and prepared antiserum against the predicted proteins. Using antisera against Jak2, we demonstrate that IL-3 stimulation results in the rapid and specific tyrosine phosphorylation of Jak2 and activates its in vitro kinase activity.
- Published
- 1993
- Full Text
- View/download PDF
48. JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin.
- Author
-
Witthuhn BA, Quelle FW, Silvennoinen O, Yi T, Tang B, Miura O, and Ihle JN
- Subjects
- 3T3 Cells, Animals, Cell Division, Enzyme Activation, Janus Kinase 2, Mice, Models, Biological, Phosphorylation, Receptors, Erythropoietin genetics, Recombinant Fusion Proteins metabolism, Transfection, Tyrosine metabolism, Erythropoietin metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins, Receptors, Erythropoietin metabolism, Signal Transduction
- Abstract
Erythropoietin (EPO) regulates the proliferation and differentiation of erythroid cells through interaction with its receptor (EPOR). Although EPOR is a member of the cytokine receptor superfamily and lacks a kinase domain, EPO induces tyrosine phosphorylation, which is correlated with gene transcription and mitogenesis. Here we demonstrate that EPO induces tyrosine phosphorylation of JAK2 kinase and activates its in vitro autophosphorylation. Using EPOR mutants, phosphorylation and activation of kinase activity correlate with the induction of mitogenesis. Furthermore, JAK2 physically associates with a membrane-proximal region of the EPOR cytoplasmic domain that is required for biological activity. The results support the hypothesis that JAK2 is the kinase that couples EPO binding to tyrosine phosphorylation and mitogenesis.
- Published
- 1993
- Full Text
- View/download PDF
49. Cytokine-induced phosphorylation of pp100 in FDC-ER cells is at tyrosine residues.
- Author
-
Quelle FW, Egerton M, Samelson LE, and Wojchowski DM
- Subjects
- Animals, Cells, Cultured, Mice, Phosphorylation drug effects, Phosphotyrosine, Receptors, Antigen, T-Cell metabolism, Receptors, Erythropoietin genetics, Receptors, Erythropoietin metabolism, Tyrosine analogs & derivatives, Tyrosine isolation & purification, Cytokines pharmacology, Phosphoproteins metabolism, Tyrosine metabolism
- Abstract
Using FDC-P1 cells stably transfected with a murine erythropoietin receptor cDNA as a model, we recently have shown that erythropoietin (EPO), IL-3 and GM-CSF each induce the rapid phosphorylation of a common cytosolic target, i.e., a M(r) 100,000 phosphoprotein "pp100". Presently, we demonstrate that cytokine-induced phosphorylation of pp100 is primarily at tyrosine residues. This is shown by Western blotting with the anti-phosphotyrosine antibody PY20, and by the resistance of [32P]-pp100 to hydroxide-mediated hydrolysis of phosphates. These data, together with the recent observation by Linnekin et al. that pp100/p97 apparently associates directly with EPO receptors, suggest that pp100 may comprise an immediate common component in the signal transduction pathways of EPO, IL-3, GM-CSF and possibly other type I/II cytokine receptors. Additional analyses suggest that pp100 is distinct from a previously described M(r) 100,000 cytosolic target which is tyrosine phosphorylated in hematopoietic cells upon activation of T-cell receptors.
- Published
- 1992
- Full Text
- View/download PDF
50. Mutations in the WSAWSE and cytosolic domains of the erythropoietin receptor affect signal transduction and ligand binding and internalization.
- Author
-
Quelle DE, Quelle FW, and Wojchowski DM
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Cloning, Molecular, Culture Media, Cytosol metabolism, Endocytosis genetics, Glycosylation, Ligands, Mice, Molecular Sequence Data, Mutagenesis, Receptors, Erythropoietin chemistry, Receptors, Erythropoietin metabolism, Erythropoietin metabolism, Receptors, Erythropoietin genetics, Signal Transduction genetics
- Abstract
The terminal development of erythroid progenitor cells is promoted in part through the interaction of erythropoietin (EPO) with its cell surface receptor. This receptor and a growing family of related cytokine receptors share homologous extracellular features, including a well-conserved WSXWS motif. To explore the functional significance of this motif in the murine EPO receptor, five WSAWSE mutants were prepared and their signal-transducing, ligand binding, and endocytotic properties were compared. EPO receptors mutated at tryptophan residues (W-232, W-235----G; W-235----G; W-235----F) failed to mediate EPO-induced growth or pp100 phosphorylation, while S-236----T and E-237----K mutants exhibited partial to full activity (50 to 100% of wild-type growth and induced phosphorylation). Ligand affinity was reduced for mutant receptors (two- to fivefold), yet expression at the cell surface for all receptors was nearly equivalent. Also, the ability of mutated receptors to internalize ligand was either markedly reduced or abolished (W-235----F), indicating a role for the WSAWSE region in hormone internalization. Interestingly, receptor forms lacking 97% of the cytosolic domain (no signal-transducing capacity; binding affinity reduced two- to threefold) internalized EPO efficiently. This and all WSAWSE receptor forms studied also mediated specific cross-linking of 125I-EPO to three accessory membrane proteins (M(r)s, 120,000, 105,000, and 93,000). These findings suggest that the WSAWSE domain of the EPO receptor is important for EPO-induced signal transduction and ligand internalization. In contrast, although the cytosolic domain is required for growth signaling, it appears nonessential for efficient endocytosis.
- Published
- 1992
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.