Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell Rep ; 34(1): 108586, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406424

ABSTRACT

The cyclic GMP-AMP (cGAMP) synthase (cGAS) is a key DNA sensor that initiates STING-dependent signaling to produce type I interferons through synthesizing the secondary messenger 2'3'-cGAMP. In this study, we confirm previous studies showing that cGAS is located both in the cytoplasm and in the nucleus. Nuclear accumulation is observed when leptomycin B is used to block the exportin, CRM1 protein. As a result, leptomycin B impairs the production of interferons in response to DNA stimulation. We further identify a functional nuclear export signal (NES) in cGAS, 169LEKLKL174. Mutating this NES leads to the sequestration of cGAS within the nucleus and the loss of interferon response to cytosolic DNA treatment, and it further determines the key amino acid to L172. Collectively, our data demonstrate that the cytosolic DNA-sensing function of cGAS depends on its presence within the cytoplasm, which is warranted by a functional NES.


Subject(s)
Cytosol/metabolism , DNA/metabolism , Karyopherins/metabolism , Nuclear Export Signals , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Amino Acid Sequence , Fatty Acids, Unsaturated/pharmacology , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Interferon Type I/drug effects , Interferon Type I/metabolism , Interferon-beta/metabolism , Karyopherins/drug effects , Karyopherins/immunology , Membrane Proteins/metabolism , Mutation , Nucleotidyltransferases/immunology , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/immunology , Signal Transduction , THP-1 Cells , Exportin 1 Protein
2.
Bioorg Chem ; 103: 104162, 2020 10.
Article in English | MEDLINE | ID: mdl-32890988

ABSTRACT

In this work, 2'-alkoxymethyl substituted klavuzon derivatives were prepared starting from 2-methyl-1-naphthoic acid in eight steps. Anticancer potencies of the synthesized compounds were evaluated by performing MTT cell viability test over cancerous and healthy pancreatic cell lines, along with CRM1 inhibitory properties in HeLa cells by immunostaining and Topo I inhibition properties by supercoiled DNA relaxation assay. Their cytotoxic activities were also presented in hepatocellular carcinoma cells (HuH-7) derived 3D spheroids. Among the tested klavuzon derivatives, isobutoxymethyl substituted klavuzon showed the highest selectivity of cytotoxic activity against pancreatic cancer cell line. They showed potent Topo I inhibition while their CRM1 inhibitory properties somehow diminished compared to 4'-alkylsubstituted klavuzons. The most cytotoxic 2'-methoxymethyl derivative inhibited the growth of the spheroids derived from HuH-7 cell lines and PI staining exhibited time and concentration dependent cell death in 3D spheroids.


Subject(s)
DNA Topoisomerases, Type I/drug effects , Karyopherins/drug effects , Naphthalenes/chemistry , Naphthalenes/therapeutic use , Neoplasms/drug therapy , Pyrans/chemistry , Pyrans/therapeutic use , Receptors, Cytoplasmic and Nuclear/drug effects , Humans , Naphthalenes/pharmacology , Pyrans/pharmacology , Structure-Activity Relationship , Exportin 1 Protein
3.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30541831

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of hospitalization of infants and young children, causing considerable respiratory disease and repeat infections that may lead to chronic respiratory conditions such as asthma, wheezing, and bronchitis. RSV causes ∼34 million new episodes of lower respiratory tract illness (LRTI) in children younger than 5 years of age, with >3 million hospitalizations due to severe RSV-associated LRTI. The standard of care is limited to symptomatic relief as there are no approved vaccines and few effective antiviral drugs; thus, a safe and efficacious RSV therapeutic is needed. Therapeutic targeting of host proteins hijacked by RSV to facilitate replication is a promising antiviral strategy as targeting the host reduces the likelihood of developing drug resistance. The nuclear export of the RSV M protein, mediated by the nuclear export protein exportin 1 (XPO1), is crucial for RSV assembly and budding. Inhibition of RSV M protein export by leptomycin B correlated with reduced RSV replication in vitro In this study, we evaluated the anti-RSV efficacy of Verdinexor (KPT-335), a small molecule designed to reversibly inhibit XPO1-mediated nuclear export. KPT-335 inhibited XPO1-mediated transport and reduced RSV replication in vitro KPT-335 was effective against RSV A and B strains and reduced viral replication following prophylactic or therapeutic administration. Inhibition of RSV replication by KPT-335 was due to a combined effect of reduced XPO1 expression, disruption of the nuclear export of RSV M protein, and inactivation of the NF-κB signaling pathway.IMPORTANCE RSV is an important cause of LRTI in infants and young children for which there are no suitable antiviral drugs offered. We evaluated the efficacy of KPT-335 as an anti-RSV drug and show that KPT-335 inhibits XPO1-mediated nuclear export, leading to nuclear accumulation of RSV M protein and reduction in RSV levels. KPT-335 treatment also resulted in inhibition of proinflammatory pathways, which has important implications for its effectiveness in vivo.


Subject(s)
Acrylamides/pharmacology , Hydrazines/pharmacology , Respiratory Syncytial Viruses/drug effects , Virus Replication/drug effects , A549 Cells , Acrylamides/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Glycoproteins/immunology , Humans , Hydrazines/metabolism , Karyopherins/drug effects , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Vero Cells , Exportin 1 Protein
4.
Hepatology ; 66(2): 602-615, 2017 08.
Article in English | MEDLINE | ID: mdl-28257601

ABSTRACT

Increased inflammatory signaling by Kupffer cells contributes to alcoholic liver disease (ALD). Here we investigated the impact of small, specific-sized hyaluronic acid of 35 kD (HA35) on ethanol-induced sensitization of Kupffer cells, as well as ethanol-induced liver injury in mice. Unbiased analysis of microRNA (miRNA) expression in Kupffer cells identified miRNAs regulated by both ethanol and HA35. Toll-like receptor 4 (TLR4)-mediated signaling was assessed in primary cultures of Kupffer cells from ethanol- and pair-fed rats after treatment with HA35. Female C57BL6/J mice were fed ethanol or pair-fed control diets and treated or not with HA35. TLR4 signaling was increased in Kupffer cells by ethanol; this sensitization was normalized by ex vivo treatment with HA35. Next generation sequencing of Kupffer cell miRNA identified miRNA 181b-3p (miR181b-3p) as sensitive to both ethanol and HA35. Importin α5, a protein involved in p65 translocation to the nucleus, was identified as a target of miR181b-3p; importin α5 protein was increased in Kupffer cells from ethanol-fed rats, but decreased by HA35 treatment. Overexpression of miR181b-3p decreased importin α5 expression and normalized lipopolysaccharide-stimulated tumor necrosis factor α expression in Kupffer cells from ethanol-fed rats. In a mouse model of ALD, ethanol feeding decreased miR181b-3p in liver and increased expression of importin α5 in nonparenchymal cells. Treatment with HA35 normalized these changes and also protected mice from ethanol-induced liver and intestinal injury. CONCLUSION: miR181b-3p is dynamically regulated in Kupffer cells and mouse liver in response to ethanol and treatment with HA35. miR181b-3p modulates expression of importin α5 and sensitivity of TLR4-mediated signaling. This study identifies a miR181b-3p-importin α5 axis in regulating inflammatory signaling pathways in hepatic macrophages. (Hepatology 2017;66:602-615).


Subject(s)
Ethanol/pharmacology , Karyopherins/genetics , Liver Diseases, Alcoholic/metabolism , MicroRNAs/metabolism , Toll-Like Receptor 4/genetics , Animals , Biopsy, Needle , Cells, Cultured , Disease Models, Animal , Ethanol/adverse effects , Female , Gene Expression Regulation , History, 18th Century , Immunohistochemistry , Karyopherins/drug effects , Kupffer Cells/cytology , Kupffer Cells/metabolism , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/drug effects , Random Allocation , Rats , Rats, Wistar , Reference Values , Signal Transduction
5.
J Hematol Oncol ; 10(1): 47, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28196522

ABSTRACT

Many recent publications highlight the large role of the pivotal eukaryotic nuclear export protein exportin-1 (XPO1) in the oncogenesis of several malignancies, and there is emerging evidence that XPO1 inhibition is a key target against cancer. The clinical validation of the pharmacological inhibition of XPO1 was recently achieved with the development of the selective inhibitor of nuclear export compounds, displaying an interesting anti-tumor activity in patients with massive pre-treated hematological malignancies. Recent reports have shown molecular alterations in the gene encoding XPO1 and showed a mutation hotspot (E571K) in the following two hematological malignancies with similar phenotypes and natural histories: primary mediastinal diffuse large B cell lymphoma and classical Hodgkin's lymphoma. Emerging evidence suggests that the mutant XPO1 E571K plays a role in carcinogenesis, and this variant is quantifiable in tumor and plasma cell-free DNA of patients using highly sensitive molecular biology techniques, such as digital PCR and next-generation sequencing. Therefore, it was proposed that the XPO1 E571K variant may serve as a minimal residual disease tool in this setting. To clarify and summarize the recent findings on the role of XPO1 in B cell hematological malignancies, we conducted a literature search to present the major publications establishing the landscape of XPO1 molecular alterations, their impact on the XPO1 protein, their interest as biomarkers, and investigations into the development of new XPO1-targeted therapies in B cell hematological malignancies.


Subject(s)
Hematologic Neoplasms/pathology , Karyopherins/drug effects , Molecular Targeted Therapy , Mutation , Receptors, Cytoplasmic and Nuclear/drug effects , B-Lymphocytes/pathology , Carcinogenesis , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hodgkin Disease , Humans , Karyopherins/genetics , Lymphoma, Large B-Cell, Diffuse , Receptors, Cytoplasmic and Nuclear/genetics , Exportin 1 Protein
6.
Leukemia ; 27(12): 2357-65, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23752175

ABSTRACT

RNA interference screening identified XPO1 (exportin 1) among the 55 most vulnerable targets in multiple myeloma (MM). XPO1 encodes CRM1, a nuclear export protein. XPO1 expression increases with MM disease progression. Patients with MM have a higher expression of XPO1 compared with normal plasma cells (P<0.04) and to patients with monoclonal gammopathy of undetermined significance/smoldering MM (P<0.0001). The highest XPO1 level was found in human MM cell lines (HMCLs). A selective inhibitor of nuclear export compound KPT-276 specifically and irreversibly inhibits the nuclear export function of XPO1. The viability of 12 HMCLs treated with KTP-276 was significantly reduced. KPT-276 also actively induced apoptosis in primary MM patient samples. In gene expression analyses, two genes of probable relevance were dysregulated by KPT-276: cell division cycle 25 homolog A (CDC25A) and bromodomain-containing protein 4 (BRD4), both of which are associated with c-MYC pathway. Western blotting and reverse transcription-PCR confirm that c-MYC, CDC25A and BRD4 are all downregulated after treatment with KPT-276. KPT-276 reduced monoclonal spikes in the Vk*MYC transgenic MM mouse model, and inhibited tumor growth in a xenograft MM mouse model. A phase I clinical trial of an analog of KPT-276 is ongoing in hematological malignancies including MM.


Subject(s)
Acrylamides/pharmacology , Biological Transport/drug effects , Cell Nucleus/drug effects , Genome-Wide Association Study , Karyopherins/genetics , Multiple Myeloma/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/pharmacology , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Profiling , Humans , Karyopherins/drug effects , Mice , RNA Interference , Receptors, Cytoplasmic and Nuclear/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays , Exportin 1 Protein
7.
J Urol ; 189(6): 2317-26, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23079374

ABSTRACT

PURPOSE: Renal cell carcinoma often presents asymptomatically and patients are commonly diagnosed at the metastatic stage, when treatment options are limited and survival is poor. Since progression-free survival using current therapy for metastatic renal cell carcinoma is only 1 to 2 years and existing drugs are associated with a high resistance rate, new pharmacological targets are needed. We identified and evaluated the nuclear exporter protein CRM1 as a novel potential therapy for renal cell carcinoma. MATERIALS AND METHODS: We tested the efficacy of the CRM1 inhibitors KPT-185 and 251 in several renal cell carcinoma cell lines and in a renal cell carcinoma xenograft model. Apoptosis and cell cycle arrest were quantified and localization of p53 family proteins was assessed using standard techniques. RESULTS: KPT-185 attenuated CRM1 and showed increased cytotoxicity in renal cell carcinoma cells in vitro with evidence of increased apoptosis as well as cell cycle arrest. KPT-185 caused p53 and p21 to remain primarily in the nucleus in all renal cell carcinoma cell lines, suggesting that the mechanism of action of these compounds depends on tumor suppressor protein localization. Furthermore, when administered orally in a high grade renal cell carcinoma xenograft model, the bioavailable CRM1 inhibitor KPT-251 significantly inhibited tumor growth in vivo with the expected on target effects and no obvious toxicity. CONCLUSIONS: The CRM1 inhibitor protein family is a novel therapeutic target for renal cell carcinoma that deserves further intensive investigation for this and other urological malignancies.


Subject(s)
Acrylates/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Karyopherins/drug effects , Kidney Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Receptors, Cytoplasmic and Nuclear/drug effects , Triazoles/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Cell Line, Tumor/drug effects , Disease Models, Animal , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Karyopherins/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Male , Mice , Mice, Nude , Neoplasm Transplantation , Random Allocation , Receptors, Cytoplasmic and Nuclear/genetics , Sensitivity and Specificity , Survival Rate , Tumor Burden/drug effects , Exportin 1 Protein
8.
Nat Chem Biol ; 8(2): 179-84, 2011 Dec 25.
Article in English | MEDLINE | ID: mdl-22198731

ABSTRACT

In biology-oriented synthesis, the scaffolds of biologically relevant compound classes inspire the synthesis of focused compound collections enriched in bioactivity. This criterion is, in particular, met by the scaffolds of natural products selected in evolution. The synthesis of natural product-inspired compound collections calls for efficient reaction sequences that preferably combine multiple individual transformations in one operation. Here we report the development of a one-pot, twelve-step cascade reaction sequence that includes nine different reactions and two opposing kinds of organocatalysis. The cascade sequence proceeds within 10-30 min and transforms readily available substrates into complex indoloquinolizines that resemble the core tetracyclic scaffold of numerous polycyclic indole alkaloids. Biological investigation of a corresponding focused compound collection revealed modulators of centrosome integrity, termed centrocountins, which caused fragmented and supernumerary centrosomes, chromosome congression defects, multipolar mitotic spindles, acentrosomal spindle poles and multipolar cell division by targeting the centrosome-associated proteins nucleophosmin and Crm1.


Subject(s)
Biological Products/chemical synthesis , Centrosome/drug effects , Alkaloids/chemical synthesis , Alkaloids/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Karyopherins/drug effects , Nuclear Proteins/drug effects , Nucleophosmin , Quinolizines/chemical synthesis , Quinolizines/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Exportin 1 Protein
9.
Mol Cell Biol ; 30(2): 470-80, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19917721

ABSTRACT

Muscle atrophy is a debilitating process associated with many chronic wasting diseases, like cancer, diabetes, sepsis, and renal failure. Rapid loss of muscle mass occurs mainly through the activation of protein breakdown by the ubiquitin proteasome pathway. Foxo3a transcription factor is critical for muscle atrophy, since it activates the expression of ubiquitin ligase Atrogin-1. In several models of atrophy, inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway induces nuclear import of Foxo3a through an Akt-dependent process. This study aimed to identify signaling pathways involved in the control of Foxo3a nuclear translocation in muscle cells. We observed that after nuclear import of Foxo3a by PI3K/Akt pathway inhibition, activation of stress-activated protein kinase (SAPK) pathways induced nuclear export of Foxo3a through CRM1. This mechanism involved the c-Jun NH(2)-terminal kinase (JNK) signaling pathway and was independent of Akt. Likewise, we showed that inhibition of p38 induced a massive nuclear relocalization of Foxo3a. Our results thus suggest that SAPKs are involved in the control of Foxo3a nucleocytoplasmic translocation in C2C12 cells. Moreover, activation of SAPKs decreases the expression of Atrogin-1, and stable C2C12 myotubes, in which the p38 pathway is constitutively activated, present partial protection against atrophy.


Subject(s)
Forkhead Transcription Factors/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle Cells/metabolism , Muscular Atrophy/metabolism , Animals , Anthracenes/pharmacology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromones/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/drug effects , Humans , Isoquinolines/pharmacology , Karyopherins/drug effects , Karyopherins/metabolism , MAP Kinase Kinase 3/drug effects , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase 4/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Morpholines/pharmacology , Muscle Cells/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/drug effects , Muscle Proteins/metabolism , Muscular Atrophy/genetics , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , SKP Cullin F-Box Protein Ligases/drug effects , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Transfection , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Exportin 1 Protein
10.
Cancer ; 112(8): 1733-43, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18306389

ABSTRACT

BACKGROUND: The human nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 (CRM1) mediates the nuclear export of proteins and messenger RNAs and, thus, is an important regulator of subcellular distribution of key molecules. Whereas cell-biologic studies have suggested a fundamental role for CRM1 in the regulation of mitosis, the expression of this protein in human tumor tissue has not been investigated to date. METHODS: In this study, the expression of CRM1 was analyzed in a cohort of 88 ovarian tumors and 12 ovarian cell lines for the first time to the authors' knowledge. RESULTS: Immunohistochemistry revealed increased nuclear (52.7%) and cytoplasmic (56.8%) expression of CRM1 in 74 carcinomas compared with the expression revealed in borderline tumors and benign lesions. Similarly, CRM1 expression was increased in ovarian cancer cell lines compared with human ovarian surface epithelial cells. Cytoplasmic CRM1 expression was related significantly to advanced tumor stage (P= .043), poorly differentiated carcinomas (P= .011), and higher mitotic rate (P= .008). Nuclear CRM1 was associated significantly with cyclooxygenase-2 (COX-2) expression (P= .002) and poor overall survival (P= .01). Because it was demonstrated previously that blocking of CRM1 by leptomycin B (LMB) contributes to the inhibition of nuclear export, the authors used a set of mechanistic assays to study the effects of CRM1 inhibition in cancer cells. Treatment of OVCAR-3 cells with LMB revealed a significant reduction of cell proliferation and increased apoptosis as well as suppressed interleukin-1beta-induced COX-2 expression. CONCLUSIONS: The current results indicated that CRM1 is expressed in a subpopulation of ovarian carcinomas with aggressive behavior and is related to poor patient outcome. A correlation also was demonstrated between CRM1 and COX-2 expression in ovarian cancer tissue. Furthermore, the treatment of ovarian cancer cells with LMB revealed a reduction in COX-2 expression. Therefore, the authors suggest that CRM1 may be an interesting biomarker for the assessment of patient prognosis and a molecular target for anticancer treatment.


Subject(s)
Karyopherins/analysis , Ovarian Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/analysis , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Carcinoma/pathology , Cell Line, Tumor , Cell Nucleus/ultrastructure , Cell Proliferation/drug effects , Cohort Studies , Cyclooxygenase 2/analysis , Cytoplasm/ultrastructure , Epithelial Cells/pathology , Fatty Acids, Unsaturated/therapeutic use , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-1beta/drug effects , Karyopherins/drug effects , Middle Aged , Mitosis/genetics , Neoplasm Staging , Prognosis , Receptors, Cytoplasmic and Nuclear/drug effects , Survival Rate , Exportin 1 Protein
11.
J Virol ; 81(8): 4298-304, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17202223

ABSTRACT

The nucleocapsid (N) protein of infectious bronchitis virus (IBV) localizes to the cytoplasm and nucleolus and contains an eight-amino-acid nucleolar retention motif. In this study, a leucine-rich nuclear export signal (NES) (291-LQLDGLHL-298) present in the C-terminal region of the IBV N protein was analyzed by using alanine substitution and deletion mutagenesis to investigate the relative contributions that leucine residues make to nuclear export and where these residues are located on the structure of the IBV N protein. The analysis indicated that Leu296 and Leu298 are required for efficient nuclear export of the protein. Structural information indicated that both of these amino acids are available for interaction with protein complexes involved in this process. However, export of N protein from the nucleus/nucleolus was not inhibited by leptomycin B treatment, indicating that N protein nuclear export is independent of the CRM1-mediated export pathway.


Subject(s)
Infectious bronchitis virus/physiology , Nuclear Export Signals , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Amino Acid Substitution , Animals , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , Karyopherins/drug effects , Microscopy, Fluorescence , Models, Molecular , Mutagenesis, Site-Directed , Receptors, Cytoplasmic and Nuclear/drug effects , Sequence Deletion , Vero Cells , Exportin 1 Protein
12.
J Leukoc Biol ; 81(3): 741-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17135572

ABSTRACT

High mobility group box 1 (HMGB1) can be actively secreted by macrophages/monocytes in response to exogenous and endogenous inflammatory stimuli (such as bacterial endotoxin, TNF-alpha, IL-1, and IFN-gamma) or passively released by necrotic cells and mediates innate and adaptive inflammatory responses to infection and injury. Here, we demonstrated that a reactive oxygen species, hydrogen peroxide (H(2)O(2)), induces active and passive HMGB1 release from macrophage and monocyte cultures in a time- and dose-dependent manner. At nontoxic doses (e.g., 0.0125-0.125 mM), H(2)O(2) induced HMGB1 cytoplasmic translocation and active release within 3-24 h. At higher concentrations (e.g., 0.25 mM), however, H(2)O(2) exhibited cytotoxicity to macrophage and monocyte cell cultures and consequently, triggered active and passive HMGB1 release. In addition, H(2)O(2) stimulated potential interaction of HMGB1 with a nuclear export factor, chromosome region maintenance (CRM1), in macrophage/monocyte cultures. Inhibitors specific for the JNK (SP600125) and MEK (PD98059), but not p38 MAPK (SB203580), abrogated H(2)O(2)-induced, active HMGB1 release. Together, these data establish an important role for oxidative stress in inducing active HMGB1 release, potentially through a MAPK- and CRM1-dependent mechanism.


Subject(s)
HMGB1 Protein/metabolism , Hydrogen Peroxide/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Animals , Anthracenes/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Flavonoids/pharmacology , HMGB1 Protein/drug effects , Humans , Karyopherins/drug effects , Karyopherins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Monocytes/immunology , Monocytes/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Reactive Oxygen Species/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Time Factors , Exportin 1 Protein
13.
FEBS Lett ; 576(1-2): 27-30, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15474004

ABSTRACT

Research on the export of proteins and nucleic acids from the nucleus to the cytoplasm has greatly gained from the discovery that the actinobacterial toxin leptomycin B (LMB) specifically inactivates the export receptor chromosomal region maintenance 1 (CRM1). Recently, it was shown that myxobacterial cytotoxins, named ratjadones (RATs), also bind to CRM1 and inhibit nuclear export. However, the reaction mechanism of RATs was not resolved. Here, we show that LMB and RAT A employ the same molecular mechanism to inactivate CRM1. Alkylation of residue Cys528 of CRM1 determines both LMB and RAT sensitivity and prevents nuclear export of CRM1 cargo proteins.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antifungal Agents/pharmacology , Fatty Acids, Unsaturated/pharmacology , Karyopherins/antagonists & inhibitors , Pyrones/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Active Transport, Cell Nucleus/physiology , Alkylation , Amino Acid Substitution , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cysteine/chemistry , Glutathione Transferase/metabolism , HeLa Cells , Humans , Karyopherins/chemistry , Karyopherins/drug effects , Karyopherins/genetics , Karyopherins/metabolism , Microinjections , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Exportin 1 Protein
14.
Exp Cell Res ; 299(1): 209-26, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15302588

ABSTRACT

Elucidating dynamic aspects of intracellular localization of proteins is essential to decipher their functional interaction networks. Although transcription factors lacking a detectable cytoplasmic fraction have been generally considered compartmentalized in the nucleus, some were found to shuttle into the cytoplasm, suggesting functional interactions therein. To further investigate how common, specific and quantitative is this traffic, we have employed the heterokaryon assay for a small-scale survey of nuclear factors not previously tested for their nucleo-cytoplasmic motion. We show that a subset of cAMP response element (CRE) binding proteins of the CREM type shuttles within a biologically meaningful time frame, revealing a continuous flow into the cytoplasm that persists during signaling. Their dynamic behavior, not involving the classical Exportin-1 pathway, could be ascribed to C-terminal sequences, containing, in addition to the bZIP domain and the NLS, a nuclear export activity and an inhibitory activity at an adjacent site. Other proteins examined in this study either did not shuttle significantly or, like CREB and distinct CREM isoforms, shuttled with markedly delayed kinetics, denoting considerable selectivity of this traffic. These findings raise the possibility that events associated with bi-directional transport and periodic transit through the cytoplasm may modulate activities of select nuclear transcription factors.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear , Repressor Proteins/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Cricetinae , Cyclic AMP Response Element Modulator , Cytoplasm/genetics , DNA-Binding Proteins/genetics , Fatty Acids, Unsaturated/pharmacology , HeLa Cells , Humans , Karyopherins/drug effects , Karyopherins/genetics , Karyopherins/metabolism , Mice , NIH 3T3 Cells , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Exportin 1 Protein
15.
J Biol Chem ; 277(35): 32310-9, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12065584

ABSTRACT

The dioxin receptor is a ligand-dependent transcription factor that mediates the biological effects of dioxin and related environmental pollutants. In the absence of ligand the receptor is present in the cytoplasmic compartment of the cell associated with the hsp90-dependent chaperone complex. This complex regulates several functions of the receptor such as ligand binding and nuclear import. Furthermore, intracellular localization of the receptor is modulated by multiple factors such as the export protein CRM-1 and the hsp90-associated immunophilin XAP-2. We have identified the mechanism of XAP-2-induced cytoplasmic localization of the receptor and studied the potential cross-talk between CRM-1 and XAP-2. We show that XAP-2 anchors the ligand-free receptor to cytoskeletal structures. This effect is blocked upon treatment with the actin inhibitor cytochalasin B, whereas the tubulin inhibitor colchicine had no effect on receptor localization. In addition, we show that the receptor interacts with CRM-1 both in the presence and absence of ligand. CRM-1-mediated nuclear export occurs independently of XAP-2. Our data provide evidence that CRM-1 and XAP-2 act in parallel through different mechanisms and target different interfaces of the receptor. These results suggest that two pathways cooperate to localize the non-activated receptor in the cytoplasmic compartment of the cell.


Subject(s)
Receptors, Aryl Hydrocarbon/metabolism , Receptors, Cytoplasmic and Nuclear , Colchicine/pharmacology , Cytochalasin B/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , Green Fluorescent Proteins , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Karyopherins/drug effects , Karyopherins/metabolism , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Chaperones/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Protein Biosynthesis , Proteins/drug effects , Proteins/metabolism , Receptors, Aryl Hydrocarbon/genetics , Recombinant Fusion Proteins/metabolism , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...