Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Transfus Clin Biol ; 20(2): 144-7, 2013 May.
Article in French | MEDLINE | ID: mdl-23643330

ABSTRACT

Erythropoiesis is finely regulated by two major cytokines, stem cell factor (SCF) and erythropoietin (Epo). Decrease levels of Epo result in caspase activation and erythroid progenitors apoptosis. However, normal erythroid cell maturation requests caspase activation and cleavage of various caspase substrates, except the erythroid transcription factor GATA-1, that is protected by interaction with the chaperone HSP70 in the nucleus. Therefore, molecular abnormalities associated with decrease of HSP70 expression in the nucleus may result in ineffective erythropoiesis characterized by apoptosis and impaired maturation of erythroid precursors. These findings open new potential targeted therapies for erythroid disorders.


Subject(s)
Erythroblasts/cytology , Erythropoiesis/physiology , HSP70 Heat-Shock Proteins/physiology , Animals , Apoptosis , Caspases/physiology , Cell Differentiation , Cell Nucleus/metabolism , Enzyme Activation , Erythrocyte Aging , Erythropoietin/physiology , GATA1 Transcription Factor/metabolism , Humans , Myelodysplastic Syndromes/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Protein Processing, Post-Translational , Proteolysis , Stem Cell Factor/physiology , Thalassemia/metabolism
2.
Cell Death Differ ; 18(4): 678-89, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21072057

ABSTRACT

Stem cell factor (SCF) and erythropoietin are strictly required for preventing apoptosis and stimulating proliferation, allowing the differentiation of erythroid precursors from colony-forming unit-E to the polychromatophilic stage. In contrast, terminal maturation to generate reticulocytes occurs independently of cytokine signaling by a mechanism not fully understood. Terminal differentiation is characterized by a sequence of morphological changes including a progressive decrease in cell size, chromatin condensation in the nucleus and disappearance of organelles, which requires transient caspase activation. These events are followed by nucleus extrusion as a consequence of plasma membrane and cytoskeleton reorganization. Here, we show that in early step, SCF stimulates the Rho/ROCK pathway until the basophilic stage. Thereafter, ROCK-1 is activated independently of Rho signaling by caspase-3-mediated cleavage, allowing terminal maturation at least in part through phosphorylation of the light chain of myosin II. Therefore, in this differentiation system, final maturation occurs independently of SCF signaling through caspase-induced ROCK-1 kinase activation.


Subject(s)
Caspase 3/metabolism , Cytokines/metabolism , Erythroblasts/cytology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Cell Differentiation , Cell Size , Chromatin/physiology , Erythroblasts/enzymology , Erythroblasts/metabolism , Humans , Myosin Type II/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cell Factor/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
3.
Cell Mol Life Sci ; 65(7-8): 1123-32, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18193168

ABSTRACT

CYLD is a protein with tumor suppressor properties which was originally discovered associated with cylindromatosis, an inherited cancer exclusively affecting the folicullo-sebaceous-apocrine unit of the epidermis. CYLD exhibits deubiquitinating activity and acts as a negative regulator of NF-kappaB and JNK signaling through its interaction with NEMO and TRAF2. Recent data suggest that this is unlikely to be its unique function in vivo. CYLD has also been shown to control other seemingly disparate cellular processes, such as proximal T cell receptor signaling, TrkA endocytosis and mitosis. In each case, this enzyme appears to act by regulating a specific type of polyubiquitination, K63 polyubiquitination, that does not result in recognition and degradation of proteins by the proteasome but instead controls their activity through diverse mechanisms.


Subject(s)
NF-kappa B/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Genetic Diseases, Inborn/metabolism , Humans , Immunity , JNK Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
4.
Cell Death Differ ; 15(1): 152-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932497

ABSTRACT

Apoptosis is mediated by cysteine-dependent, aspartate-directed proteases of the caspase family that proteolyse strategic intracellular substrates to induce cell suicide. We describe here that engagement of apoptotic processes by Fas triggering or by staurosporine stimulation leads to the caspase-dependent inactivation of the nuclear factor kappa B (NF-kappaB) pathway after cleavage of IKK1 (IkappaB kinase 1) and NEMO (NF-kappaB essential modulator), which are needed to transduce NF-kappaB activation signals. In this study, we have analyzed in more detail, the role of NEMO cleavage, as NEMO, but not IKK1, is important for the pro-survival actions of NF-kappaB. We demonstrate that NEMO is cleaved after Asp355 to remove the last 64 C-terminal amino acids. This short form was unable to rescue NF-kappaB activation by tumor necrosis factor-alpha (TNF-alpha) when transfected in NEMO-deficient cells. Consequently, inactivation of NEMO resulted in an inhibition of the expression of antiapoptotic NF-kappaB-target genes coding for caspase inhibitors (cIAP-1, cIAP-2) or adaptors of the TNF receptor family. NEMO-deficient Jurkat cells transiently expressing a non-cleavable mutant of NEMO were less sensitive to TNF-alpha-induced apoptosis. Therefore, downmodulation of NF-kappaB activation via the proteolytic cleavage of NEMO could represent an amplification loop for apoptosis.


Subject(s)
Apoptosis , Caspases/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis/physiology , Cell Survival , Enzyme Activation , Humans , Jurkat Cells , Signal Transduction
6.
Hum Mutat ; 28(7): 703-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17354266

ABSTRACT

Anhidrotic ectodermal dysplasia (EDA) is a disorder of ectodermal differentiation characterized by sparse hair, abnormal or missing teeth, and inability to sweat. X-linked EDA is the most common form, caused by mutations in the EDA gene, which encodes ectodysplasin, a member of the tumor necrosis factor (TNF) family. Autosomal dominant and recessive forms of EDA have been also described and are accounted for by two genes. Mutations in EDAR, encoding a TNF receptor (EDAR) cause both dominant and recessive forms. In addition, mutations in a recently identified gene, EDARADD, encoding EDAR-associated death domain (EDARADD) have been shown to cause autosomal recessive EDA. Here, we report a large Moroccan family with an autosomal dominant EDA. We mapped the disease gene to chromosome 1q42.2-q43, and identified a novel missense mutation in the EDARADD gene (c.335T>G, p.Leu112Arg). Thus, the EDARADD gene accounts for both recessive and dominant EDA. EDAR is activated by its ligand, ectodysplasin, and uses EDARADD to build an intracellular complex and activate nuclear factor kappa B (NF-kB). We compared the functional consequences of the dominant (p.Leu112Arg) and recessive mutation (p.Glu142Lys), which both occurred in the death domain (DD) of EDARADD. We demonstrated that the p.Leu112Arg mutation completely abrogated NF-kB activation, whereas the p.Glu142Lys retained the ability to significantly activate the NF-kB pathway. The p.Leu112Arg mutation is probably a dominant negative form as its cotransfection impaired the wild-type EDARADD's ability to activate NF-kB. Our results confirm that NF-kB activation is impaired in EDA and support the role of EDARADD DD as a downstream effector of EDAR signaling.


Subject(s)
Ectodermal Dysplasia 1, Anhidrotic/genetics , Edar Receptor/genetics , Genes, Dominant , Base Sequence , DNA Primers , Female , Humans , Male , Mutation , NF-kappa B/metabolism , Pedigree , Phenotype
7.
Oncogene ; 25(51): 6831-43, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17072331

ABSTRACT

The nuclear factor-kappa B (NF-kappaB) signaling pathway is a multi-component pathway that regulates the expression of hundreds of genes that are involved in diverse and key cellular and organismal processes, including cell proliferation, cell survival, the cellular stress response, innate immunity and inflammation. Not surprisingly, mis-regulation of the NF-kappaB pathway, either by mutation or epigenetic mechanisms, is involved in many human and animal diseases, especially ones associated with chronic inflammation, immunodeficiency or cancer. This review describes human diseases in which mutations in the components of the core NF-kappaB signaling pathway have been implicated and discusses the molecular mechanisms by which these alterations in NF-kappaB signaling are likely to contribute to the disease pathology. These mutations can be germline or somatic and include gene amplification (e.g., REL), point mutations and deletions (REL, NFKB2, IKBA, CYLD, NEMO) and chromosomal translocations (BCL-3). In addition, human genetic diseases are briefly described wherein mutations affect protein modifiers or transducers of NF-kappaB signaling or disrupt NF-kappaB-binding sites in promoters/enhancers.


Subject(s)
Genetic Diseases, Inborn/metabolism , Mutation , NF-kappa B/metabolism , Signal Transduction , Humans
8.
Cell Death Differ ; 13(5): 843-51, 2006 May.
Article in English | MEDLINE | ID: mdl-16397577

ABSTRACT

The recent identification of genetic diseases (incontinentia pigmenti, anhidrotic ectodermal dysplasia with immunodeficiency and cylindromatosis) resulting from mutations affecting components of the nuclear factor-kappaB (NF-kappaB) signaling pathway provides a unique opportunity to understand the function of NF-kappaB in vivo. Besides confirming the importance of NF-kappaB in innate and acquired immunity or bone mass control, analysis of these diseases has uncovered new critical roles played by this transcription factor in the development and homeostasis of the epidermis and the proper function of lymphatic vessels. In addition, the identified mutations will help understanding at the molecular level how NF-kappaB is activated in response to cell stimulation.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Animals , Deubiquitinating Enzyme CYLD , Gene Expression Regulation , I-kappa B Kinase/genetics , I-kappa B Proteins/genetics , Interleukin-1 Receptor-Associated Kinases , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
Pathol Biol (Paris) ; 51(1): 27-32, 2003 Feb.
Article in French | MEDLINE | ID: mdl-12628289

ABSTRACT

The t(1;22)(p13;q13) translocation is specifically associated with infant acute megakaryoblastic leukemia (M7). We have recently characterized the two genes involved in this translocation: OTT (One Two Two) and MAL (Megakaryoblastic Acute Leukemia) respectively located on chromosome 1 and 22. The t(1;22) translocation results in the fusion of these genes in all the cases studied to date. We summarize here present knowledge regarding this translocation.


Subject(s)
Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 22 , Leukemia, Megakaryoblastic, Acute/genetics , Translocation, Genetic , Humans
11.
Nat Genet ; 27(3): 277-85, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11242109

ABSTRACT

The molecular basis of X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) has remained elusive. Here we report hypomorphic mutations in the gene IKBKG in 12 males with EDA-ID from 8 kindreds, and 2 patients with a related and hitherto unrecognized syndrome of EDA-ID with osteopetrosis and lymphoedema (OL-EDA-ID). Mutations in the coding region of IKBKG are associated with EDA-ID, and stop codon mutations, with OL-EDA-ID. IKBKG encodes NEMO, the regulatory subunit of the IKK (IkappaB kinase) complex, which is essential for NF-kappaB signaling. Germline loss-of-function mutations in IKBKG are lethal in male fetuses. We show that IKBKG mutations causing OL-EDA-ID and EDA-ID impair but do not abolish NF-kappaB signaling. We also show that the ectodysplasin receptor, DL, triggers NF-kappaB through the NEMO protein, indicating that EDA results from impaired NF-kappaB signaling. Finally, we show that abnormal immunity in OL-EDA-ID patients results from impaired cell responses to lipopolysaccharide, interleukin (IL)-1beta, IL-18, TNFalpha and CD154. We thus report for the first time that impaired but not abolished NF-kappaB signaling in humans results in two related syndromes that associate specific developmental and immunological defects.


Subject(s)
Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Adolescent , Child , Child, Preschool , Codon, Terminator/genetics , Ectodermal Dysplasia/metabolism , Ectodysplasins , Genetic Linkage , Humans , I-kappa B Kinase , Immunity, Cellular , Immunologic Deficiency Syndromes/metabolism , Infant , Male , Membrane Proteins/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Syndrome , X Chromosome/genetics
12.
Am J Hum Genet ; 68(3): 765-71, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179023

ABSTRACT

Familial incontinentia pigmenti (IP [MIM 308310]), or Bloch-Sulzberger syndrome, is an X-linked dominant and male-lethal disorder. We recently demonstrated that mutations in NEMO (IKK-gamma), which encodes a critical component of the NF-kappaB signaling pathway, were responsible for IP. Virtually all mutations eliminate the production of NEMO, causing the typical skewing of X inactivation in female individuals and lethality in male individuals, possibly through enhanced sensitivity to apoptosis. Most mutations also give rise to classic signs of IP, but, in this report, we describe two mutations in families with atypical phenotypes. Remarkably, each family included a male individual with unusual signs, including postnatal survival and either immune dysfunction or hematopoietic disturbance. We found two duplication mutations in these families, at a cytosine tract in exon 10 of NEMO, both of which remove the zinc (Zn) finger at the C-terminus of the protein. Two deletion mutations were also identified in the same tract in additional families. However, only the duplication mutations allowed male individuals to survive, and affected female individuals with duplication mutations demonstrated random or slight skewing of X inactivation. Similarly, NF-kappaB activation was diminished in the presence of duplication mutations and was completely absent in cells with deletion mutations. These results strongly indicate that male individuals can also suffer from IP caused by NEMO mutations, and we therefore urge a reevaluation of the diagnostic criteria.


Subject(s)
Carrier Proteins , Cytosine , Exons , Incontinentia Pigmenti/genetics , Mitogen-Activated Protein Kinases/genetics , Amino Acid Sequence , Base Sequence , Chromosome Aberrations , Female , Humans , I-kappa B Kinase , Incontinentia Pigmenti/enzymology , Male , Molecular Sequence Data , Pedigree , Phenotype , Sequence Deletion , X Chromosome
13.
J Biol Chem ; 276(12): 8898-903, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11114307

ABSTRACT

Following challenge with proinflammatory stimuli or generation of DNA double strand breaks (DSBs), transcription factor NF-kappaB translocates from the cytoplasm to the nucleus to activate expression of target genes. In addition, NF-kappaB plays a key role in protecting cells from proapoptotic stimuli, including DSBs. Patients suffering from the genetic disorder ataxia-telangiectasia, caused by mutations in the ATM gene, are highly sensitive to inducers of DSBs, such as ionizing radiation. Similar hypersensitivity is displayed by cell lines derived from ataxia-telangiectasia patients or Atm knockout mice. The ATM protein, a member of the phosphatidylinositol 3-kinase (PI3K)-like family, is a multifunctional protein kinase whose activity is stimulated by DSBs. As both ATM and NF-kappaB deficiencies result in increased sensitivity to DSBs, we examined the role of ATM in NF-kappaB activation. We report that ATM is essential for NF-kappaB activation in response to DSBs but not proinflammatory stimuli, and this activity is mediated via the IkappaB kinase complex. DNA-dependent protein kinase, another member of the PI3K-like family, PI3K itself, and c-Abl, a nuclear tyrosine kinase, are not required for this response.


Subject(s)
DNA Damage , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Androstadienes/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , I-kappa B Kinase , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins , Wortmannin
14.
J Chromatogr A ; 881(1-2): 599-606, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10905739

ABSTRACT

Furosine and furfural products of the Maillard reaction are used as specific indicators of the effect of heating treatments on milk quality. Their contents were measured in representative samples of store- and name-brand ultra-high-temperature-treated milks using RP-HPLC with UV detection. Furosine contents ranged from 40.32 to 50.67 and from 65.48 to 310.58 mg/100 g protein in name- and store-brand milks, respectively. Of the furfurals, only hydroxymethylfurfural was detected. The free hydroxymethylfurfural contents of store-brand milks ranged from 0.22 to 1.70 mg/100 g protein. Total hydroxymethylfurfural contents ranged from 0.29 to 0.41 and from 0.72 to 2.21 mg/100 g protein, for name- and store-brands, respectively.


Subject(s)
Chromatography, High Pressure Liquid/methods , Maillard Reaction , Milk/chemistry , Animals , Cattle , Hot Temperature , Spectrophotometry, Ultraviolet
15.
Mol Cell ; 5(6): 981-92, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10911992

ABSTRACT

Disruption of the X-linked gene encoding NF-kappa B essential modulator (NEMO) produces male embryonic lethality, completely blocks NF-kappa B activation by proinflammatory cytokines, and interferes with the generation and/or persistence of lymphocytes. Heterozygous female mice develop patchy skin lesions with massive granulocyte infiltration and hyperproliferation and increased apoptosis of keratinocytes. Diseased animals present severe growth retardation and early mortality. Surviving mice recover almost completely, presumably through clearing the skin of NEMO-deficient keratinocytes. Male lethality and strikingly similar skin lesions in heterozygous females are hallmarks of the human genetic disorder incontinentia pigmenti (IP). Together with the recent discovery that mutations in the human NEMO gene cause IP, our results indicate that we have created a mouse model for that disease.


Subject(s)
Incontinentia Pigmenti/genetics , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Cell Division , Cells, Cultured , Chemotaxis, Leukocyte , Cytokines/metabolism , Cytokines/pharmacology , Disease Models, Animal , Female , Gene Targeting , Heterozygote , Humans , Hyperpigmentation , I-kappa B Kinase , Incontinentia Pigmenti/embryology , Incontinentia Pigmenti/enzymology , Incontinentia Pigmenti/pathology , Keratinocytes/enzymology , Keratinocytes/pathology , Liver/embryology , Liver/pathology , Male , Melanins/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Skin/embryology , Skin/enzymology , Skin/metabolism , Skin/pathology , Up-Regulation
16.
Nature ; 405(6785): 466-72, 2000 May 25.
Article in English | MEDLINE | ID: mdl-10839543

ABSTRACT

Familial incontinentia pigmenti (IP; MIM 308310) is a genodermatosis that segregates as an X-linked dominant disorder and is usually lethal prenatally in males. In affected females it causes highly variable abnormalities of the skin, hair, nails, teeth, eyes and central nervous system. The prominent skin signs occur in four classic cutaneous stages: perinatal inflammatory vesicles, verrucous patches, a distinctive pattern of hyperpigmentation and dermal scarring. Cells expressing the mutated X chromosome are eliminated selectively around the time of birth, so females with IP exhibit extremely skewed X-inactivation. The reasons for cell death in females and in utero lethality in males are unknown. The locus for IP has been linked genetically to the factor VIII gene in Xq28 (ref. 3). The gene for NEMO (NF-kappaB essential modulator)/IKKgamma (IkappaB kinase-gamma) has been mapped to a position 200 kilobases proximal to the factor VIII locus. NEMO is required for the activation of the transcription factor NF-kappaB and is therefore central to many immune, inflammatory and apoptotic pathways. Here we show that most cases of IP are due to mutations of this locus and that a new genomic rearrangement accounts for 80% of new mutations. As a consequence, NF-kappaB activation is defective in IP cells.


Subject(s)
Gene Rearrangement , Incontinentia Pigmenti/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Exons , Female , Humans , I-kappa B Kinase , Incontinentia Pigmenti/embryology , Male , Molecular Sequence Data , Mutation , NF-kappa B/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction
17.
J Biol Chem ; 275(30): 22780-9, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10807909

ABSTRACT

The NF-kappaB signaling pathway plays a crucial role in the immune, inflammatory, and apoptotic responses. Recently, we identified the NF-kappaB Essential Modulator (NEMO) as an essential component of this pathway. NEMO is a structural and regulatory subunit of the high molecular kinase complex (IKK) responsible for the phosphorylation of NF-kappaB inhibitors. Data base searching led to the isolation of a cDNA encoding a protein we called NRP (NEMO-related protein), which shows a strong homology to NEMO. Here we show that NRP is present in a novel high molecular weight complex, that contains none of the known members of the IKK complex. Consistently, we could not observe any effect of NRP on NF-kappaB signaling. Nonetheless, we could demonstrate that treatment with phorbol esters induces NRP phosphorylation and decreases its half-life. This phosphorylation event could only be inhibited by K-252a and stauroporin. We also show that de novo expression of NRP can be induced by interferon and tumor necrosis factor alpha and that these two stimuli have a synergistic effect on NRP expression. In addition, we observed that endogenous NRP is associated with the Golgi apparatus. Analogous to NEMO, we find that NRP is associated in a complex with two kinases, suggesting that NRP could play a similar role in another signaling pathway.


Subject(s)
Gene Expression Regulation/drug effects , Interferons/pharmacology , NF-kappa B/metabolism , Phosphoproteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor TFIIIA , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Sequence , Animals , Carbazoles/pharmacology , Cell Cycle Proteins , Cell Line , Golgi Apparatus/metabolism , Humans , Indole Alkaloids , Membrane Transport Proteins , Mice , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Sequence Homology, Amino Acid , Staurosporine/pharmacology
18.
Cell Signal ; 12(11-12): 759-68, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11152962

ABSTRACT

Inactive nuclear factor kappaB (NF-kappaB) complexes are retained in the cytoplasm by binding to inhibitory proteins, such as IkappaBalpha. Various stimuli lead to phosphorylation and subsequent processing of IkappaBalpha in the 26S proteasome and import of the active NF-kappaB transcription factor into the nucleus. In agreement with our previous finding that p90(rsk1) is essential for TPA-induced activation of NF-kappaB in Adenovirus 5E1-transformed Baby Rat Kidney cells, we now report that the MEK/ERK/p90(rsk1) inhibitor U0126 efficiently blocks TPA-induced IkappaBalpha processing in these cells. However, in U2OS cells, the cytokine-inducible IkappaB kinase complex (IKK) is the essential component of the TPA signal transduction pathway. Activation of the IKK complex in response to TPA is mediated by PKC-alpha, since both the PKC inhibitor GF109203 and a catalytically inactive PKC-alpha mutant inhibit activation of endogenous IKK by TPA, but not by tumor necrosis factor-alpha (TNF-alpha). We conclude that IKK is an integrator of TNF-alpha and TPA signal transduction pathways in U2OS cells.


Subject(s)
I-kappa B Proteins , Isoenzymes/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Butadienes/pharmacology , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Genes, Dominant/genetics , Humans , I-kappa B Kinase , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Kinetics , MAP Kinase Kinase 1 , Maleimides/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/drug effects , Mutation/genetics , NF-KappaB Inhibitor alpha , Nitriles/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
19.
Sci STKE ; 2000(58): pe1, 2000 Nov 14.
Article in English | MEDLINE | ID: mdl-11752619

ABSTRACT

The components of the nuclear factor-kappaB (NF-kappaB) family of transcription factors are critical for regulating the response to immune challenges. Recently, a role for NF-kappaB in skin biology has been revealed. Within the cascade of proteins whose activities impinge upon the activation of NF-kappaB, the NEMO (NF-kappaB essential modulator)/IKKgamma protein is required for the activation of the IkappaB kinases, which in turn, promote the degradation of IkappaB proteins, leading to the derepression of NF-kappaB activity. Courtois and Israël discuss the role of NEMO/IKKgamma in normal physiological activation of NF-kappaB and the consequences of defective NF-kappaB activation, as an effect of NEMO/IKKgamma mutations, which can lead to incontinentia pigmenti, a disease marked by alopecia, tooth eruption, skin lesions, and changes in skin pigmentation.


Subject(s)
Incontinentia Pigmenti/enzymology , NF-kappa B/deficiency , NF-kappa B/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Humans , I-kappa B Kinase , Incontinentia Pigmenti/genetics , NF-kappa B/genetics , Protein Serine-Threonine Kinases/genetics
20.
Cell ; 93(7): 1231-40, 1998 Jun 26.
Article in English | MEDLINE | ID: mdl-9657155

ABSTRACT

We have characterized a flat cellular variant of HTLV-1 Tax-transformed rat fibroblasts, 5R, which is unresponsive to all tested NF-kappaB activating stimuli, and we report here its genetic complementation. The recovered full-length cDNA encodes a 48 kDa protein, NEMO (NF-kappaB Essential MOdulator), which contains a putative leucine zipper motif. This protein is absent from 5R cells, is part of the high molecular weight IkappaB kinase complex, and is required for its formation. In vitro, NEMO can homodimerize and directly interacts with IKK-2. The NEMO cDNA was also able to complement another NF-kappaB-unresponsive cell line, 1.3E2, in which the protein is also absent, allowing us to demonstrate that this factor is required not only for Tax but also for LPS, PMA, and IL-1 stimulation of NF-kappaB activity.


Subject(s)
DNA-Binding Proteins/physiology , Genetic Complementation Test , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/chemistry , Transcription Factors/physiology , Transcriptional Activation/physiology , Amino Acid Sequence , Animals , Cell Line, Transformed , Cloning, Molecular/methods , DNA, Complementary/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Fibroblasts , Gene Products, tax , Human T-lymphotropic virus 1 , I-kappa B Kinase , Leucine Zippers , Molecular Sequence Data , Protein Serine-Threonine Kinases/biosynthesis , Rats , Sequence Analysis, DNA , Transcription Factors/chemistry , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...