Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Med (Lausanne) ; 11: 1364038, 2024.
Article in English | MEDLINE | ID: mdl-38695031

ABSTRACT

Delayed post-hypoxic leukoencephalopathy (DPHL) is a poorly recognized syndrome characterized by neuropsychiatric symptoms following recovery from an acute hypoxic episode. Although most cases are related to carbon monoxide poisoning, some have been linked to excessive opioid use. Opioid intoxication has recently become known for manifesting the characteristic imaging findings involving cerebellar, hippocampal, and basal nuclei transient edema with restricted diffusion (CHANTER) syndrome. Herein, we present a patient with severe disturbances in consciousness who was initially diagnosed with CO poisoning but was later found to have taken excessive tramadol. Magnetic resonance imaging (MRI) in the acute phase revealed abnormal intensities in the bilateral globus pallidus and the cerebellum, indicative of CHANTER syndrome. After intensive care, his level of consciousness was restored. However, around the 3rd week after hospitalization, his consciousness gradually deteriorated and he developed severe neurological symptoms. Another MRI on day 25 revealed a new diffuse white matter abnormality; DPHL was suspected. Cerebrospinal fluid collected on day 28 revealed significantly elevated myelin basic protein levels. Although it was challenging to decide on a treatment plan, hyperbaric oxygen (HBO) therapy trials were initiated on day 58; the patient's condition improved after a series of HBO sessions. MRI revealed gradual shrinkage of the white matter abnormality. A total of 63 consecutive HBO sessions were performed, leading to the successful resolution of the serious neurological symptoms. While the effectiveness of HBO therapy for DPHL remains inconclusive, especially in opioid-related cases, this patient made a remarkable recovery, likely due to the therapeutic effect of improved cerebral blood flow and oxygenation.

2.
Cells ; 13(3)2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38334661

ABSTRACT

Murine leukemia viruses (MuLVs) are simple retroviruses that cause several diseases in mice. Retroviruses encode three basic genes: gag, pol, and env. Gag is translated as a polyprotein and moves to assembly sites where viral particles are shaped by cleavage of poly-Gag. Viral release depends on the intracellular trafficking of viral proteins, which is determined by both viral and cellular factors. ADP-ribosylation factor 6 (Arf6) is a small GTPase that regulates vesicular trafficking and recycling of different types of cargo in cells. Arf6 also activates phospholipase D (PLD) and phosphatidylinositol-4-phosphate 5-kinase (PIP5K) and produces phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2). We investigated how Arf6 affected MuLV release with a constitutively active form of Arf6, Arf6Q67L. Expression of Arf6Q67L impaired Gag release by accumulating Gag at PI(4,5)P2-enriched compartments in the cytoplasm. Treatment of the inhibitors for PLD and PIP5K impaired or recovered MuLV Gag release in the cells expressing GFP (control) and Arf6Q67L, implying that regulation of PI(4,5)P2 through PLD and PIP5K affected MuLV release. Interference with the phosphoinositide 3-kinases, mammalian target of rapamycin (mTOR) pathway, and vacuolar-type ATPase activities showed further impairment of Gag release from the cells expressing Arf6Q67L. In contrast, mTOR inhibition increased Gag release in the control cells. The proteasome inhibitors reduced viral release in the cells regardless of Arf6Q67L expression. These data outline the differences in MuLV release under the controlled and overactivated Arf6 conditions and provide new insight into pathways for MuLV release.


Subject(s)
ADP-Ribosylation Factor 6 , Leukemia Virus, Murine , Viral Proteins , Animals , Mice , Leukemia Virus, Murine/physiology , TOR Serine-Threonine Kinases
3.
Biochem Biophys Res Commun ; 500(2): 365-369, 2018 06 02.
Article in English | MEDLINE | ID: mdl-29660337

ABSTRACT

Murine leukemia virus (MuLV) is a retrovirus known causing leukemia and neurological disorders in mice, and its viral life cycle and pathogenesis have been investigated extensively over the past decades. As a natural antiviral agent, betulinic acid is a pentacyclic triterpenoid that can be found in the bark of several species of plants (particularly the white birch). One of the hurdles for betulinic acid to release its antiviral potency is its poor water solubility. In this study, we synthesized more water-soluble ionic derivatives of betulinic acid, and examined their activities against Moloney MuLV (M-MuLV). The mouse fibroblast cells stably infected with M-MuLV, 43D cells, were treated with various doses of betulinic acids and its derivatives, and the viral structural protein Gag in cells and media were detected by western blots. Two ionic derivatives containing the benzalkonium cation were found to inhibit the virus production into media and decreased Gag in cells. However, a cell proliferation assay showed that the benzalkonium cation inhibited the growth of 43D cells, suggesting that our ionic derivatives limited virus production through the inhibition of metabolism in 43D cells. Interestingly, all of these betulinic acid compounds exhibited a minimum impact on the processing and release of Gag from 43D cells, which outlines the differences of viral maturation between MuLV and human immunodeficiency virus.


Subject(s)
Moloney murine leukemia virus/physiology , Triterpenes/pharmacology , Virus Replication/drug effects , Animals , Benzalkonium Compounds/pharmacology , Cell Death/drug effects , Cell Line , Ions , Mice , Moloney murine leukemia virus/drug effects , Pentacyclic Triterpenes , Triterpenes/chemistry , Betulinic Acid
4.
Retrovirology ; 12: 68, 2015 Aug 08.
Article in English | MEDLINE | ID: mdl-26253512

ABSTRACT

BACKGROUND: Koala retrovirus (KoRV) is an endogenous and exogenous retrovirus of koalas that may cause lymphoma. As for many other gammaretroviruses, the KoRV genome can potentially encode an alternate form of Gag protein, glyco-gag. RESULTS: In this study, a convenient assay for assessing KoRV infectivity in vitro was employed: the use of DERSE cells (initially developed to search for infectious xenotropic murine leukemia-like viruses). Using infection of DERSE and other human cell lines (HEK293T), no evidence for expression of glyco-gag by KoRV was found, either in expression of glyco-gag protein or changes in infectivity when the putative glyco-gag reading frame was mutated. Since glyco-gag mediates resistance of Moloney murine leukemia virus to the restriction factor APOBEC3, the sensitivity of KoRV (wt or putatively mutant for glyco-gag) to restriction by murine (mA3) or human APOBEC3s was investigated. Both mA3 and hA3G potently inhibited KoRV infectivity. Interestingly, hA3G restriction was accompanied by extensive G → A hypermutation during reverse transcription while mA3 restriction was not. Glyco-gag status did not affect the results. CONCLUSIONS: These results indicate that the mechanisms of APOBEC3 restriction of KoRV by hA3G and mA3 differ (deamination dependent vs. independent) and glyco-gag does not play a role in the restriction.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine Deaminase/metabolism , Gammaretrovirus/physiology , Gene Products, gag/metabolism , Phascolarctidae/virology , Virus Replication , APOBEC Deaminases , Amino Acid Sequence , Animals , Gammaretrovirus/genetics , Gammaretrovirus/pathogenicity , Gene Products, gag/chemistry , Gene Products, gag/genetics , HEK293 Cells , Humans , Mice , Open Reading Frames , Reverse Transcription , Sequence Alignment
5.
J Virol ; 88(12): 7054-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672043

ABSTRACT

UNLABELLED: Previous studies showed that short hairpin RNA (shRNA) knockdown of the RNA lariat debranching enzyme (DBR1) led to a decrease in the production of HIV-1 cDNA. To further characterize this effect, DBR1 shRNA was introduced into GHOST-R5X4 cells, followed by infection at a multiplicity near unity with HIV-1 or an HIV-1-derived vector. DNA and RNA were isolated from whole cells and from cytoplasmic and nuclear fractions at different times postinfection. Inhibition of DBR1 had little or no effect on the formation of minus-strand strong-stop cDNA but caused a significant reduction in the formation of intermediate and full-length cDNA. Moreover, minus-strand strong-stop DNA rapidly accumulated in the cytoplasm in the first 2 h of infection but shifted to the nuclear fraction by 6 h postinfection. Regardless of DBR1 inhibition, greater than 95% of intermediate-length and full-length HIV-1 cDNA was found in the nuclear fraction at all time points. Thus, under these experimental conditions, HIV-1 cDNA synthesis was initiated in the cytoplasm and completed in the nucleus or perinuclear region of the infected cell. When nuclear import of the HIV-1 reverse transcription complex was blocked by expressing a truncated form of the mRNA cleavage and polyadenylation factor CPSF6, the completion of HIV-1 vector cDNA synthesis was detected in the cytoplasm, where it was not inhibited by DBR1 knockdown. Refinement of the cell fractionation procedure indicated that the completion of reverse transcription occurred both within nuclei and in the perinuclear region. Taken together the results indicate that in infections at a multiplicity near 1, HIV-1 reverse transcription is completed in the nucleus or perinuclear region of the infected cell, where it is dependent on DBR1. When nuclear transport is inhibited, reverse transcription is completed in the cytoplasm in a DBR1-independent manner. Thus, there are at least two mechanisms of HIV-1 reverse transcription that require different factors and occur in different intracellular locations. IMPORTANCE: This study shows that HIV-1 reverse transcription starts in the cytoplasm but is completed in or on the surface of the nucleus. Moreover, we show that nuclear reverse transcription is dependent on the activity of the human RNA lariat debranchng enzyme (DBR1), while cytoplasmic reverse transcription is not. These findings may provide new avenues for inhibiting HIV-1 replication and therefore may lead to new medicines for treating HIV-1-infected individuals.


Subject(s)
DNA, Complementary/genetics , HIV Infections/enzymology , HIV Infections/virology , HIV-1/genetics , RNA Nucleotidyltransferases/genetics , Cell Nucleus/virology , Cytoplasm/virology , DNA, Complementary/metabolism , Gene Knockdown Techniques , HIV Infections/genetics , HIV-1/physiology , Humans , RNA Nucleotidyltransferases/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication
6.
Proc Natl Acad Sci U S A ; 110(22): 9078-83, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671100

ABSTRACT

Pathogenic retroviruses have evolved multiple means for evading host restriction factors such as apolipoprotein B editing complex (APOBEC3) proteins. Here, we show that murine leukemia virus (MLV) has a unique means of counteracting APOBEC3 and other cytosolic sensors of viral nucleic acid. Using virus isolated from infected WT and APOBEC3 KO mice, we demonstrate that the MLV glycosylated Gag protein (glyco-Gag) enhances viral core stability. Moreover, in vitro endogenous reverse transcription reactions of the glyco-Gag mutant virus were substantially inhibited compared with WT virus, but only in the presence of APOBEC3. Thus, glyco-Gag rendered the reverse transcription complex in the viral core resistant to APOBEC3. Glyco-Gag in the virion also rendered MLV resistant to other cytosolic sensors of viral reverse transcription products in newly infected cells. Strikingly, glyco-Gag mutant virus reverted to glyco-Gag-containing virus only in WT and not APOBEC3 KO mice, indicating that counteracting APOBEC3 is the major function of glyco-Gag. Thus, in contrast to the HIV viral infectivity factor protein, which prevents APOBEC3 packaging in the virion, the MLV glyco-Gag protein uses a unique mechanism to counteract the antiviral action of APOBEC3 in vivo--namely, protecting the reverse transcription complex in viral cores from APOBEC3. These data suggest that capsid integrity may play a critical role in virus resistance to intrinsic cellular antiviral resistance factors that act at the early stages of infection.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Gene Products, gag/metabolism , Host-Pathogen Interactions/physiology , Leukemia Virus, Murine/physiology , Reverse Transcription/physiology , Animals , Blotting, Western , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Primers/genetics , Gene Products, gag/pharmacology , Glycosylation , Leukemia Virus, Murine/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction
7.
Virus Genes ; 45(3): 508-17, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22864547

ABSTRACT

Jaagsiekte sheep retrovirus (JSRV) is the causative agent of a transmissible lung cancer in sheep. A unique feature is that JSRV envelope protein is also the oncogene for this virus. Previous studies have identified the cytoplasmic tail (CT) of the envelope transmembrane (TM) protein as critical for transformation although other regions of Env have also been implicated. In this study, the roles of other Env regions in transformation were investigated. Chimeras between JSRV Env and the Env of a related non-oncogenic endogenous retrovirus (enJSRV, 5F16) were used. A chimera containing the membrane-spanning region (MSR) of enJSRV inserted into JSRV Env showed substantially reduced transformation, indicating that the MSR plays a role in transformation. Transformation by this chimera was highly dependent on both Ras/Raf/MEK/MAPK and PI3K/Akt/mTOR signaling. A chimera containing the two amino acids in the TM ectodomain that distinguish JSRV and enJSRV showed modestly reduced transformation. Chimeras in the SU protein indicated that the amino terminal region of SU contributes to transformation, while the C-terminal part is not important. To test if Env trimerization is important for transformation, we mutated a leucine-rich sequence in the putative trimerization domain in the ectodomain of TM (Tri-M). This mutant could not transform cells and it did not oligomerize. However, Tri-M could complement a non-transforming mutant CT mutant (Y590F) so oligomerization is not necessary for at least some aspects of transformation. These experiments provide new insight into the regions and residues of JSRV Env protein necessary for oncogenic transformation.


Subject(s)
Cell Transformation, Viral , Jaagsiekte sheep retrovirus/genetics , Pulmonary Adenomatosis, Ovine/virology , Viral Envelope Proteins/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Jaagsiekte sheep retrovirus/metabolism , Leucine/genetics , Leucine/metabolism , Mice , Mutagenesis, Site-Directed , NIH 3T3 Cells , Plasmids/genetics , Plasmids/metabolism , Protein Multimerization , Protein Structure, Tertiary , Pulmonary Adenomatosis, Ovine/pathology , Radioimmunoprecipitation Assay , Sheep/virology , Structure-Activity Relationship , Viral Envelope Proteins/genetics
8.
Retrovirology ; 9: 58, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22828015

ABSTRACT

BACKGROUND: One of the unique features of gammaretroviruses is that they contain an additional extended form of Gag, glyco-gag, which initiates in the leader sequence. MuLV glyco-gag, gPr80Gag, promotes retrovirus replication and disease progression. Although virtually all infectious MuLVs encode glyco-gag, XMRV (xenotropic murine leukemia virus-related virus) lacks the classical gPr80Gag sequence. We examined XMRV to determine if its leader sequence contains glyco-gag activity, whether the presence of conventional gPr80Gag affects replication of XMRV, and we describe the evolution of glyco-gag-deficient MuLVs in Mus. RESULTS: We introduced several mutations disrupting two putative but noncanonical glyco-gag proteins in the leader sequence region in XMRV and found that those mutations did not affect virus release nor susceptibility to the antiviral activity of hA3G (human APOBEC3G). A chimeric XMRV encoding the Moloney MuLV (M-MuLV) leader sequence (MXMRV) demonstrated that M-MuLV glyco-gag facilitated MXMRV release and increased infectivity. Infectivity assays with several cell lines showed that glyco-gag increases XMRV infectivity in all cell lines tested, but the level of this increase varies in different cell lines. Because MuLV glyco-gag counteracts mouse APOBEC3, we investigated whether M-MuLV glyco-gag enhances XMRV infection by counteracting human APOBEC3. Comparison of hAPOBEC3 isoforms expressed in different cell lines indicated that hA3B was the most likely candidate for a restrictive hA3. However over-expression of hA3B showed no enhanced restriction of infection by XMRV compared to MXMRV. Endogenous MuLVs in the sequenced mouse genome were screened for canonical glyco-gag, which was identified in two clades of xenotropic MuLVs (X-MuLVs) and ecotropic MuLVs, but not in other X-MuLVs or in any polytropic MuLVs. CONCLUSIONS: M-MuLV glyco-gag facilitates XMRV replication, and the leader sequence region in XMRV does not encode proteins equivalent to M-MuLV glyco-gag. The fact that the ability of glyco-gag to enhance XMRV infection varies in different cell lines suggests a glyco-gag sensitive restrictive factor that further reduces XMRV infectivity. The M-MuLV glyco-gag enhancement for XMRV replication is through a hAPOBEC3 independent mechanism. The absence of glyco-gag in MuLVs carried by western European mice suggests that loss of this sequence is a relatively recent event with limited subspecies distribution.


Subject(s)
Cytosine Deaminase/metabolism , Gene Products, gag/metabolism , Glycoproteins/metabolism , Moloney murine leukemia virus/metabolism , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , APOBEC Deaminases , Amino Acid Sequence , Animals , Base Sequence , Cytidine Deaminase , Cytosine Deaminase/antagonists & inhibitors , Cytosine Deaminase/genetics , Evolution, Molecular , Gene Products, gag/classification , Gene Products, gag/genetics , Genome, Viral , Glycoproteins/genetics , Glycosylation , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Moloney murine leukemia virus/genetics , Mutagenesis, Site-Directed , Mutation , Phylogeny , Rats , Virus Release , Xenotropic murine leukemia virus-related virus/genetics , Xenotropic murine leukemia virus-related virus/metabolism
9.
mBio ; 2(1): e00341-10, 2011.
Article in English | MEDLINE | ID: mdl-21343359

ABSTRACT

Murine leukemia viruses (MuLVs) encode two forms of Gag polyprotein: the precursor for the viral core proteins (Pr65(gag) for Moloney MuLV [M-MuLV]) and a longer glycosylated form (glyco-gag, or gPr80(gag)). gPr80(gag) is translated from the same unspliced viral RNA as Pr65(gag), from an upstream in-frame CUG initiation codon. As a result, gPr80(gag) contains 88 unique N-terminal amino acids that include a signal peptide that conducts gPr80(gag) into the rough endoplasmic reticulum, where it is glycosylated, exported to the cell surface, and cleaved into two proteins of 55 and 40 kDa. The amino-terminal 55-kDa protein remains cell associated with the 88 unique amino acids exposed to the cytosol. We previously showed that gPr80(gag) facilitates efficient M-MuLV release through lipid rafts. In this report, we found that the unique N-terminal domain of gPr80(gag) is sufficient to facilitate enhanced M-MuLV particle release from transfected 293T cells. A search for cellular proteins involved in gPr80(gag) function led to cellular La protein. Overexpression of mouse or human La enhanced M-MuLV particle release in the absence of glyco-gag, and the released virus had a reduced buoyant density characteristic of increased cholesterol content. Moreover, small interfering RNA (siRNA) knockdown of human La abolished glyco-gag enhancement of M-MuLV release. These results implicate La as a cellular protein involved in M-MuLV glyco-gag function. We also found that overexpression of mouse or human La could enhance HIV-1 release in the absence of gPr80(gag). Therefore, M-MuLV and HIV-1 may share a pathway for release through lipid rafts involving La.


Subject(s)
Autoantigens/metabolism , Gene Products, gag/metabolism , Leukemia Virus, Murine/physiology , Membrane Lipids/metabolism , Peptide Fragments/metabolism , Retroviridae Infections/metabolism , Virus Shedding , Animals , Autoantigens/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Gene Products, gag/chemistry , Gene Products, gag/genetics , Glycosylation , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Humans , Leukemia Virus, Murine/chemistry , Leukemia Virus, Murine/genetics , Mice , NIH 3T3 Cells , Peptide Fragments/genetics , Protein Structure, Tertiary , Retroviridae Infections/virology
10.
Exp Ther Med ; 2(1): 89-93, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22977475

ABSTRACT

Human T-cell leukemia virus type-1 (HTLV-1) causes adult T-cell leukemia and HTLV-1-associated myelo-pathy/tropical spastic paraparesis. HTLV-1 is mainly transmitted through blood transfusion and breastfeeding, but viral proliferation in the body in vivo shortly after transmission is not well understood. To investigate whether the route of infection influences the early stages of viral proliferation, we inoculated BALB/c mice with MT-2 cells, an HTLV-1-producing human T-cell line, via different routes, and evaluated the proviral load and humoral immune responses. One month after infection, the provirus was detected in most organs of the mice infected intraperitoneally, and substantial proviral loads were detected in the peripheral blood and secondary lymphoid organs. In contrast, the mice infected intravenously and orally showed low proviral loads, and the provirus distribution was limited to the spinal cord among the intravenously inoculated mice and to the liver among the perorally inoculated mice. Mice infected intraperitoneally also exhibited higher interleukin-2 production than the mice infected intravenously or orally, or than the uninfected control mice, while anti-HTLV-1 antibody titers were comparable between the mice infected intraperitoneally and intravenously. These results demonstrate that the route of primary HTLV-1 infection influences the establishment of HTLV-1-infected cell proliferation and the cell reservoir in mice.

11.
Proc Natl Acad Sci U S A ; 107(3): 1190-5, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080538

ABSTRACT

Murine leukemia viruses encode a unique form of Gag polyprotein, gPr80gag or glyco-gag. Translation of this protein is initiated from full-length viral mRNA at an upstream initiation site in the same reading frame as Pr65(gag), the precursor for internal structural (Gag) proteins. Whereas gPr80gag is evolutionarily conserved among gammaretroviruses, its mechanism of action has been unclear, although it facilitates virus production at a late assembly or release step. Here, it is shown that gPr80gag facilitates release of Moloney murine leukemia virus (M-MuLV) from cells along an IFN-sensitive pathway. In particular, gPr80gag-facilitated release occurs through lipid rafts, because gPr80gag-negative M-MuLV has a lower cholesterol content, is less sensitive to inhibition of release by the cholesterol-depleting agent MbetaCD, and there is less Pr65gag associated with detergent-resistant membranes in mutant-infected cells. gPr80gag can also facilitate the release of HIV-1-based vector particles from human 293T cells.


Subject(s)
Cholesterol/metabolism , Gene Products, gag/metabolism , Interferons/metabolism , Leukemia Virus, Murine/metabolism , Cell Line , Glycosylation , Humans , Leukemia Virus, Murine/physiology , Microscopy, Confocal , Protein Transport
12.
J Virol ; 83(23): 12483-98, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776124

ABSTRACT

Retroviruses express Gag and Pol proteins by translation of unspliced genome-length viral RNA. For some retroviruses, transport of unspliced viral RNA to the cytoplasm is mediated by small regulatory proteins such as human immunodeficiency virus Rev, while other retroviruses contain constitutive transport elements in their RNAs that allow transport without splicing. In this study, we found that the betaretrovirus Jaagsiekte sheep retrovirus (JSRV) encodes within the env gene a trans-acting factor (Rej) necessary for the synthesis of Gag protein from unspliced viral RNA. Deletion of env sequences from a JSRV proviral expression plasmid (pTN3) abolished its ability to produce Gag polyprotein in transfected 293T cells, and Gag synthesis could be restored by cotransfection of an env expression plasmid (DeltaGP). Deletion analysis localized the complementing activity (Rej) to the putative Env signal peptide, and a signal peptide expression construct showed Rej activity. Two other betaretroviruses, mouse mammary tumor virus (MMTV) and human endogenous retrovirus type K, encode analogous factors (Rem and Rec, respectively) that are encoded from doubly spliced env mRNAs. Reverse transcriptase-PCR cloning and sequencing identified alternate internal splicing events in the 5' end of JSRV env that could signify analogous doubly spliced Rej mRNAs, and cDNA clones expressing two of them also showed Rej activity. The predicted Rej proteins contain motifs similar to those found in MMTV Rem and other analogous retroviral regulatory proteins. Interestingly, in most cell lines, JSRV expression plasmids with Rej deleted showed normal transport of unspliced JSRV RNA to the cytoplasm; however, in 293T cells Rej modestly enhanced export of unspliced viral RNA (2.8-fold). Metabolic labeling experiments with [(35)S]methionine indicated that JSRV Rej is required for the synthesis of viral Gag polyprotein. Thus, in most cell lines, the predominant function of Rej is to facilitate translation of unspliced viral mRNA.


Subject(s)
Carrier Proteins/physiology , Gene Products, gag/biosynthesis , Jaagsiekte sheep retrovirus/physiology , Viral Envelope Proteins/physiology , Virus Replication , Animals , Cell Line , Endogenous Retroviruses/genetics , Humans , Mammary Tumor Virus, Mouse/genetics , Molecular Sequence Data , Proviruses/genetics , Sequence Analysis, DNA , Sequence Deletion , Sequence Homology
13.
J Virol ; 83(23): 12499-511, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776134

ABSTRACT

Jaagsiekte sheep retrovirus (JSRV) is a simple betaretrovirus causing a contagious lung cancer of sheep. JSRV encodes unspliced and spliced viral RNAs, among which unspliced RNA encodes Gag and Pol proteins and a singly spliced mRNA encodes Env protein. In another study we found that JSRV encodes a regulatory protein, Rej, that is responsible for synthesis of Gag polyprotein from unspliced viral RNA. Rej is encoded in the 5' end of env, and it enhances nuclear export or accumulation of cytoplasmic unspliced viral RNA in 293T cells but not in most other cell lines (A. Hofacre, T. Nitta, and H. Fan, J. Virol. 83:12483-12498, 2009). In this study, we found that mutations in the 3' end of env in the context of a cytomegalovirus-driven full-length JSRV expression construct abolished Gag protein synthesis and released viruses in 293T cells. These mutants also showed deficits in accumulation of unspliced viral RNA in the cytoplasm. These mutants defined a Rej-responsive element (RejRE). Inhibition of CRM1 but not Tap function prevented nuclear export/accumulation of cytoplasmic unspliced RNA in 293T cells, similarly to other complex retroviruses that express analogous regulator proteins (e.g., human immunodeficiency virus Rev). Structural modeling of the RejRE with Zuker M-fold indicated a region with a predicted stable secondary structure. Mutational analysis in this region indicated the importance of both secondary structures and primary nucleotide sequences in a central stem-bulge-stem structure. In contrast to 293T cells, mutations in the RejRE did not affect the levels of cytoplasmic unspliced RNA in 293 cells, although the unspliced RNA showed partial degradation, perhaps due to lack of translation. RejRE-containing RNA relocalized Rej protein from the nucleus to the cytoplasm in 293 and rat 208F cells, suggesting binding of Rej to the RejRE.


Subject(s)
Carrier Proteins/metabolism , Gene Products, gag/biosynthesis , Jaagsiekte sheep retrovirus/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , Response Elements , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Humans , Jaagsiekte sheep retrovirus/genetics , Mutagenesis, Site-Directed , Protein Structure, Secondary , Rats
14.
Int J Oncol ; 35(4): 701-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19724905

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) is characterized by monoclonal proliferation of tumor cells that harbor integrated human T-cell leukemia virus type-1 (HTLV-1). These malignant cells accumulate in various organs including the liver, spleen and skin in addition to blood and lymph nodes. Although there have been several reports of animal models of HTLV-1 infection in which proviral distribution has been examined, clonal expansion of the experimentally infected host cells has not been extensively analyzed. Here we provide experimental evidence that clonal proliferation of the infected host cells occurs in the spleen for more than one year. During a 15 month period of persistent infection, two out of ten mice developed spontaneous tumors. Although the tumors were not ATL-like, cells exhibiting mono- or oligoclonal proliferation and having the same site of HTLV-1 integration were identified in tumor tissues as well as in the spleen. Quantitative analysis of the cells belonging to each cell clone suggested that these proliferating cell clones were associated with the tumors and that spontaneous tumor tissues might provide a suitable microenvironment for proliferation and accumulation of infected cell clones at the late stage of infection.


Subject(s)
Cell Proliferation , Cell Transformation, Viral , HTLV-I Infections/virology , Human T-lymphotropic virus 1/pathogenicity , Neoplasms/virology , Spleen/virology , Tumor Virus Infections/virology , Animals , Animals, Newborn , Cell Line , Clone Cells , HTLV-I Infections/pathology , Histiocytoma/pathology , Histiocytoma/virology , Humans , Leukemia, Myeloid/pathology , Leukemia, Myeloid/virology , Liposarcoma/pathology , Liposarcoma/virology , Mice , Mice, Inbred C3H , Neoplasms/pathology , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/virology , Spleen/pathology , Splenic Neoplasms/pathology , Splenic Neoplasms/virology , Time Factors , Tumor Virus Infections/pathology , Viral Load , Virus Integration
15.
Cancer Sci ; 99(5): 872-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18312467

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) causes adult T-cell leukemia and inflammatory disorders. Aberrant activation of nuclear factor-kappaB (NF-kappaB) has been linked to HTLV-1 pathogenesis and to various kinds of cancers, including adult T-cell leukemia. NF-kappaB-inducing kinase (NIK) is critical for non-canonical activation of NF-kappaB and for the development of lymphoid organs. HTLV-1 activates NF-kappaB by the non-canonical pathway, but examination of the role of NIK in proliferation of HTLV-1-infected cells in vivo has been hindered by lack of a suitable animal model. Alymphoplasia (aly/aly) mice bear a mutation of NIK, resulting in defects in the development of lymphoid organs and severe deficiencies in both humoral and cell-mediated immunity. In the present study we therefore used a mouse model of HTLV-1 infection with aly/aly mice. The number of HTLV-1-infected cells in the reservoir organs in aly/aly mice was significantly smaller than in the control group 1 month after infection. In addition, aly/aly mice did not maintain provirus for 1 year and antibodies against HTLV-1 were undetectable. These results demonstrate that the absence of functional NIK impairs primary HTLV-1 proliferation and abolishes the maintenance of provirus. Interestingly, clonal proliferation of HTLV-1-infected mouse cells was not detected in aly/aly mice, which is consistent with the lack of HTLV-1 persistence. These observations imply that the clonal proliferation of HTLV-1-infected cells in secondary lymphoid organs might be important for HTLV-1 persistence.


Subject(s)
HTLV-I Infections/enzymology , Human T-lymphotropic virus 1/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Cell Proliferation , Cell Transformation, Viral , HTLV-I Infections/virology , Humans , Lymph Nodes/metabolism , Mice , Models, Animal , Protein Serine-Threonine Kinases/genetics , RNA, Viral/metabolism , NF-kappaB-Inducing Kinase
16.
Cancer Sci ; 97(9): 836-41, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16805820

ABSTRACT

Centrosomes play pivotal roles in cell polarity, regulation of the cell cycle and chromosomal segregation. Centrosome amplification was recently described as a possible cause of aneuploidy in certain solid tumors and leukemias. ATL is a T-cell malignancy caused by HTLV-1. Although the precise mechanism of cell transformation is unclear, the HTLV-1-encoded protein, Tax, is thought to play a crucial role in leukemogenesis. Here we demonstrate that lymphocytes isolated from patients with ATL show centrosome amplification and that a human T cell line shows centrosome amplification after induction of Tax, which was suppressed by CDK inhibitors. Micronuclei formation was also observed after centrosome amplification in Tax-induced human T cells. These findings suggest that Tax deregulates CDK activity and induces centrosome amplification, which might be associated with cellular transformation by HTLV-1 and chromosomal instability in HTLV-1-infected human T cells.


Subject(s)
Cell Transformation, Viral/genetics , Centrosome/physiology , Gene Products, tax/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , T-Lymphocytes/virology , Fluorescent Antibody Technique , Gene Products, tax/metabolism , Humans , Immunoblotting , Jurkat Cells
17.
Cancer Res ; 66(8): 4020-9, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618721

ABSTRACT

The centrosome plays a fundamental role in cell division, cell polarity, and cell cycle progression. Centrosome duplication is mainly controlled by cyclin-dependent kinase 2 (CDK2)/cyclin E and cyclin A complexes, which are inhibited by the CDK inhibitors p21Cip1 and p27Kip1. It is thought that abnormal activation of CDK2 induces centrosome amplification that is frequently observed in a wide range of aggressive tumors. We previously reported that overexpression of the oncogene MYCN leads to centrosome amplification after DNA damage in neuroblastoma cells. We here show that centrosome amplification after gamma-irradiation was caused by suppression of p27 expression in MYCN-overexpressing cells. We further show that p27-/- and p27+/- mouse embryonic fibroblasts and p27-silenced human cells exhibited a significant increase in centrosome amplification after DNA damage. Moreover, abnormal mitotic cells with amplified centrosomes were frequently observed in p27-silenced cells. In response to DNA damage, the level of p27 gradually increased in normal cells independently of the ataxia telangiectasia mutated/p53 pathway, whereas Skp2, an F-box protein component of an SCF ubiquitin ligase complex that targets p27, was reduced. Additionally, p27 levels in MYCN-overexpressing cells were restored by treatment with Skp2 small interfering RNA, indicating that down-regulation of p27 by MYCN was due to high expression of Skp2. These results suggest that the accumulation of p27 after DNA damage is required for suppression of centrosome amplification, thereby preventing chromosomal instability.


Subject(s)
Centrosome/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Damage/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Centrosome/radiation effects , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Gamma Rays , Humans , Mitosis/physiology , Mitosis/radiation effects , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
18.
World J Gastroenterol ; 11(22): 3392-7, 2005 Jun 14.
Article in English | MEDLINE | ID: mdl-15948244

ABSTRACT

AIM: To establish and characterize a new cholangiocarcinoma cell line from a patient living in the Opisthorchis viverrini (O. viverrini) endemic area of Northeast Thailand. METHODS: Fresh liver biopsy and bile specimens were obtained from a 65-year-old Thai woman with cholangiocarcinoma of the porta hepatis. After digestion, the cells were cultured in Ham's F12 media. The established cell line was then characterized for growth kinetics, cell morphology, imm-unocytochemistry and cytogenetics. Tumorigenicity of the cell line was determined by heterotransplanting in nude mice. RESULTS: The primary tumor was a poorly differentiated tubular adenocarcinoma. Examination of the bile revealed malignant cells with O. viverrini eggs. The cholangioc-arcinoma cell line KKU-100 was established 4 mo after the primary culture-population doubling time was 72 h. KKU-100 possesses compact and polygonal-shaped epithelial cells. Immunocytochemically, this cell line exhibited cytokeratin, EMA, CEA, and CA125, but not alpha-fetoprotein (AFP), CA19-9, desmin, c-met, or p53. Such protein expressions parallel those of the primary tumor. Cytogenetic analysis identified aneuploidy karyotypes with a modal chromosome number of 78 and marked chromosomal structural changes. Inoculation of KKU-100 cells into nude mice produced a transplantable, poorly differentiated aden-ocarcinoma, similar to the original tumor. CONCLUSION: KKU-100 is the first egg-proven, Opisthorchis-associated cholangiocarcinoma cell line, which should prove useful for further investigations of the tumor biology of this cancer.


Subject(s)
Bile Duct Neoplasms/parasitology , Bile Ducts, Intrahepatic/parasitology , Cell Line, Tumor , Cholangiocarcinoma/parasitology , Opisthorchiasis/complications , Aged , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/pathology , Female , Humans , Opisthorchiasis/pathology
19.
Cancer Sci ; 95(4): 306-10, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15072587

ABSTRACT

Adult T-cell leukemia (ATL) occurs after a long latent period of persistent infection by human T-cell leukemia virus type 1 (HTLV-1). However, the mechanism of oncogenesis by HTLV-1 remains to be clarified. It was reported that the incidence curve of ATL versus age was consistent with a multistage carcinogenesis model. Although HTLV-1 is an oncogenic retrovirus, a mechanism of carcinogenesis in ATL by insertional mutagenesis as one step during multistage carcinogenesis has not been considered thus far, because the exact integration sites on the chromosome have not been analyzed. Here we determined the precise HTLV-1 integration sites on the human chromosome, by taking advantage of the recently available human genome database. We isolated 25 integration sites of HTLV-1 from 23 cases of ATL. Interestingly, 13 (52%) of the integration sites were within genes, a rate significantly higher than that expected in the case of random integration (P = 0.043, chi(2) test). These results suggest that preferential integration into genes at the first infection is a characteristic of HTLV-1. However considering that some of the genes are related to the regulation of cell growth, the integration of HTLV-1 into or near growth-related genes might contribute to the clonal selection of HTLV-1-infected cells during multistage carcinogenesis of ATL.


Subject(s)
Chromosome Mapping , DNA, Neoplasm/analysis , DNA, Viral/analysis , Human T-lymphotropic virus 1/isolation & purification , Leukemia-Lymphoma, Adult T-Cell/virology , Proviruses/isolation & purification , Virus Integration , Chromosomes, Human/genetics , Chromosomes, Human/ultrastructure , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Proviruses/genetics
20.
Int J Cancer ; 107(3): 375-80, 2003 Nov 10.
Article in English | MEDLINE | ID: mdl-14506736

ABSTRACT

The liver fluke infection-associated intrahepatic cholangiocarcinoma (ICC) is a major liver cancer in Northeast Thailand. The molecular basis of this ICC is poorly understood. To address possible roles of the DNA mismatch repair (MMR) system in ICC carcinogenesis, a fluorescence-labeling PCR/laser scanning technique with high sensitivity was employed to analyze genomic instability in the nuclear DNA (nDNA) and mitochondrial DNA (mtDNA) in 24 fresh and 13 formalin-fixed, paraffin-embedded tissues of ICC and their corresponding normal parts. Microsatellite instability (MSI) was assessed in nDNA, using 12 highly polymorphic loci including 5 Bethesda markers. These loci were mainly related to major MMR genes, hMSH2 and hMLH1. Also 3 (C)n and/or (C)n(A)n repeat instability at 1 noncoding region in the displacement-loop (D-loop) and 2 coding sequences in NADH dehydrogenase subunit 1 and subunit 5 gene in mtDNA were analyzed. MSI was only detected in 1 (2.7%), 6 (16.7%), 1 (2.9%), 1 (2.9%) or 2 (6.3%) out of 37, 36, 35, 35 or 32 cases at BAT-25, D2S123, D3S1611, D11S904 or D17S250, respectively. LOH was found at D3S1298, D3S1561, D5S346 and TP53 in 4 (18.2%) out of 22, 2 (18.2%) out of 11, 6 (33.3%) out of 18 and 3 (12.5%) out of 24 informative cases, respectively. In mtDNA, none except a single case out of the 37 (2.7%) exhibited repeat sequence instability in the D-loop. We conclude that the liver fluke infection-associated ICC in Thailand is classified as low frequency MSI or microsatellite stable type and that DNA MMR system, through hMSH2 and hMLH1 gene mutations, does not play a major role in its carcinogenesis.


Subject(s)
Cholangiocarcinoma/etiology , DNA-Binding Proteins , Fascioliasis/genetics , Liver Neoplasms/etiology , Microsatellite Repeats , Adaptor Proteins, Signal Transducing , Adult , Aged , Carrier Proteins , DNA, Mitochondrial/genetics , Fascioliasis/complications , Female , Humans , Loss of Heterozygosity , Male , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Nuclear Proteins , Proto-Oncogene Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...