Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 85(15): 7863-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21593145

ABSTRACT

Nipah virus (NiV) is a recently emerged zoonotic paramyxovirus whose natural reservoirs are several species of Pteropus fruit bats. NiV provokes a widespread vasculitis often associated with severe encephalitis, with up to 75% mortality in humans. We have analyzed the pathogenesis of NiV infection, using human leukocyte cultures and the hamster animal model, which closely reproduces human NiV infection. We report that human lymphocytes and monocytes are not permissive for NiV and a low level of virus replication is detected only in dendritic cells. Interestingly, despite the absence of infection, lymphocytes could efficiently bind NiV and transfer infection to endothelial and Vero cells. This lymphocyte-mediated transinfection was inhibited after proteolytic digestion and neutralization by NiV-specific antibodies, suggesting that cells could transfer infectious virus to other permissive cells without the requirement for NiV internalization. In NiV-infected hamsters, leukocytes captured and carried NiV after intraperitoneal infection without themselves being productively infected. Such NiV-loaded mononuclear leukocytes transfer lethal NiV infection into naïve animals, demonstrating efficient virus transinfection in vivo. Altogether, these results reveal a remarkable capacity of NiV to hijack leukocytes as vehicles to transinfect host cells and spread the virus throughout the organism. This mode of virus transmission represents a rapid and potent method of NiV dissemination, which may contribute to its high pathogenicity.


Subject(s)
Leukocytes/virology , Nipah Virus/physiology , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Cricetinae , DNA Primers , Humans , Mesocricetus , RNA, Viral/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Virus Replication
2.
J Cell Sci ; 119(Pt 20): 4199-206, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16984975

ABSTRACT

Insulin-secreting pancreatic beta cells are exceptionally rich in zinc. In these cells, zinc is required for zinc-insulin crystallization within secretory vesicles. Secreted zinc has also been proposed to be a paracrine and autocrine modulator of glucagon and insulin secretion in pancreatic alpha and beta cells, respectively. However, little is known about the molecular mechanisms underlying zinc accumulation in insulin-containing vesicles. We previously identified a pancreas-specific zinc transporter, ZnT-8, which colocalized with insulin in cultured beta cells. In this paper we studied its localization in human pancreatic islet cells, and its effect on cellular zinc content and insulin secretion. In human pancreatic islet cells, ZnT-8 was exclusively expressed in insulin-producing beta cells, and colocalized with insulin in these cells. ZnT-8 overexpression stimulated zinc accumulation and increased total intracellular zinc in insulin-secreting INS-1E cells. Furthermore, ZnT-8-overexpressing cells display enhanced glucose-stimulated insulin secretion compared with control cells, only for a high glucose challenge, i.e. >10 mM glucose. Altogether, these data strongly suggest that the zinc transporter ZnT-8 is a key protein for both zinc accumulation and regulation of insulin secretion in pancreatic beta cells.


Subject(s)
Cation Transport Proteins/metabolism , Glucose/pharmacology , Insulin/metabolism , Animals , Biological Transport/drug effects , Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Expression/genetics , Glucagon/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism , Zinc/metabolism , Zinc/pharmacology , Zinc Transporter 8
3.
Diabetes ; 53(9): 2330-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331542

ABSTRACT

SLC30A8, a novel member of the zinc transporter (ZnT) family, was identified by searching the human genomic and expressed sequence tag (EST) databases with the amino acid sequence of all known human ZnT. The protein (369 amino acids) predicted from this gene, ZnT-8, contains six transmembrane domains and a histidine-rich loop between transmembrane domains IV and V, like the other ZnT proteins. We demonstrated by RT-PCR on cDNA libraries and human tissue extracts that the ZnT-8 gene is solely transcribed in the pancreas, mainly in the islets of Langerhans. The gene, named SLC30A8, was cloned and sequenced. Confocal immunofluorescence analysis revealed that a ZnT-8-EGFP (enhanced green fluorescent protein) fusion product colocalized with insulin in the secretory pathway granules of the insulin-secreting INS-1 cells. Exposure of the ZnT-8-EGFP stably expressing HeLa cells to 75 micromol/l zinc caused an accumulation of zinc in intracellular vesicles compared with cells expressing EGFP alone. These results identified ZnT-8 as a ZnT specific to the pancreas and expressed in beta-cells. Because ZnT-8 facilitates the accumulation of zinc from the cytoplasm into intracellular vesicles, ZnT-8 may be a major component for providing zinc to insulin maturation and/or storage processes in insulin-secreting pancreatic beta-cells.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Insulin/metabolism , Islets of Langerhans/physiology , Secretory Vesicles/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Green Fluorescent Proteins , HeLa Cells , Humans , Insulin Secretion , Luminescent Proteins/genetics , Molecular Sequence Data , RNA, Messenger/analysis , Zinc/metabolism , Zinc Transporter 8
4.
Biochem Pharmacol ; 68(4): 699-709, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15276077

ABSTRACT

Intracellular zinc levels are strictly regulated by zinc channels and zinc-binding proteins to maintain cellular zinc-dependent functions. We demonstrated a correlation between extracellular zinc concentration and intracellular exchangeable zinc levels using the fluorescent zinc-specific probes zinquin and zinpyr-1. The effect of extracellular zinc status on the regulation of the two trans-Golgi network directed zinc transporters ZnT-5 and ZnT-7 was next studied by real-time RT-PCR in zinc supplemented or depleted HeLa cells. While sub-toxic extracellular zinc addition strongly induced the efflux transporter ZnT-1 gene expression, consistent with its activation by the transcription factor MTF-1, treated HeLa cells did not display any change in ZnT-5 and ZnT-7 mRNA levels compared to control cells. In contrast, zinc depletion induced by non-toxic doses of the zinc chelator TPEN (N,N,N',N' tetrakis-(2 pyridylmethyl) ethylene diamine) resulted in a up to eight-fold induction of transporters ZnT-5 and ZnT-7 mRNA levels, providing the first evidence of a transcriptional control of these two zinc efflux transporters by zinc deficiency in cultured cells.


Subject(s)
Gene Expression Regulation/drug effects , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Zinc/pharmacology , Cation Transport Proteins , Cell Survival/drug effects , Ethylenediamines/pharmacology , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
BMC Genomics ; 5(1): 32, 2004 May 23.
Article in English | MEDLINE | ID: mdl-15154973

ABSTRACT

BACKGROUND: Intracellular zinc concentration and localization are strictly regulated by two main protein components, metallothioneins and membrane transporters. In mammalian cells, two membrane transporters family are involved in intracellular zinc homeostasis: the uptake transporters called SLC39 or Zip family and the efflux transporters called SLC30 or ZnT family. ZnT proteins are members of the cation diffusion facilitator (CDF) family of metal ion transporters. RESULTS: From genomic databanks analysis, we identified the full-length sequences of two novel SLC30 genes, SLC30A8 and SLC30A10, extending the SLC30 family to ten members. We used an expressed sequence tag (EST) data mining strategy to determine the pattern of ZnT genes expression in tissues. In silico results obtained for already studied ZnT sequences were compared to experimental data, previously published. We determined an overall good correlation with expression pattern obtained by RT-PCR or immunomethods, particularly for highly tissue specific genes. CONCLUSION: The method presented herein provides a useful tool to complete gene families from sequencing programs and to produce preliminary expression data to select the proper biological samples for laboratory experimentation.


Subject(s)
Multigene Family , Amino Acid Sequence , Base Sequence , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 8/genetics , Databases, Factual , Expressed Sequence Tags , Gene Expression Regulation , Genes , Humans , Molecular Sequence Data , Organ Specificity , Phylogeny , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...