Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Biol ; 207(6): 695-703, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25512390

ABSTRACT

Upon starvation, Grh1, a peripheral membrane protein located at endoplasmic reticulum (ER) exit sites and early Golgi in Saccharomyces cerevisiae under growth conditions, relocates to a compartment called compartment for unconventional protein secretion (CUPS). Here we report that CUPS lack Golgi enzymes, but contain the coat protein complex II (COPII) vesicle tethering protein Uso1 and the Golgi t-SNARE Sed5. Interestingly, CUPS biogenesis is independent of COPII- and COPI-mediated membrane transport. Pik1- and Sec7-mediated membrane export from the late Golgi is required for complete assembly of CUPS, and Vps34 is needed for their maintenance. CUPS formation is triggered by glucose, but not nitrogen starvation. Moreover, upon return to growth conditions, CUPS are absorbed into the ER, and not the vacuole. Altogether our findings indicate that CUPS are not specialized autophagosomes as suggested previously. We suggest that starvation triggers relocation of secretory and endosomal membranes, but not their enzymes, to generate CUPS to sort and secrete proteins that do not enter, or are not processed by enzymes of the ER-Golgi pathway of secretion.


Subject(s)
COP-Coated Vesicles/metabolism , Saccharomyces cerevisiae/metabolism , Secretory Vesicles/metabolism , 1-Phosphatidylinositol 4-Kinase/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Culture Media , Endoplasmic Reticulum/metabolism , Glucose/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Transport , Qa-SNARE Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/metabolism , Vesicular Transport Proteins/metabolism
2.
Elife ; 2: e00658, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23741618

ABSTRACT

Mucin 5AC (MUC5AC) is secreted by goblet cells of the respiratory tract and, surprisingly, also expressed de novo in mucus secreting cancer lines. siRNA-mediated knockdown of 7343 human gene products in a human colonic cancer goblet cell line (HT29-18N2) revealed new proteins, including a Ca(2+)-activated channel TRPM5, for MUC5AC secretion. TRPM5 was required for PMA and ATP-induced secretion of MUC5AC from the post-Golgi secretory granules. Stable knockdown of TRPM5 reduced a TRPM5-like current and ATP-mediated Ca(2+) signal. ATP-induced MUC5AC secretion depended strongly on Ca(2+) influx, which was markedly reduced in TRPM5 knockdown cells. The difference in ATP-induced Ca(2+) entry between control and TRPM5 knockdown cells was abrogated in the absence of extracellular Ca(2+) and by inhibition of the Na(+)/Ca(2+) exchanger (NCX). Accordingly, MUC5AC secretion was reduced by inhibition of NCX. Thus TRPM5 activation by ATP couples TRPM5-mediated Na(+) entry to promote Ca(2+) uptake via an NCX to trigger MUC5AC secretion. DOI:http://dx.doi.org/10.7554/eLife.00658.001.


Subject(s)
Calcium/metabolism , Colon/metabolism , Goblet Cells/metabolism , Mucins/metabolism , TRPM Cation Channels/physiology , Colon/cytology , Goblet Cells/cytology , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Humans , TRPM Cation Channels/metabolism , Tetradecanoylphorbol Acetate/pharmacology
3.
J Leukoc Biol ; 85(4): 703-10, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19116231

ABSTRACT

Multiple genetic studies in humans indicate a role for solute carrier family 11a member 1 [SLC11A1; formerly natural resistance-associated macrophage protein 1 (NRAMP1)] in autoimmune disease susceptibility, including ulcerative colitis. Murine Slc11a1 has many pleiotropic effects on macrophage activation and proinflammatory responses. To determine which of these are important in ulcerative colitis, we established a phenotype for oral dextran sulfate sodium (DSS)-induced acute colitis in congenic Slc11a1 wild-type (wt) and mutant (mt) mice on a B10 background. For over 7 days of treatment with 2% DSS in the drinking water, Slc11a1 wt mice showed enhanced acute ulcerative colitis, as demonstrated by significantly greater body weight loss and reduction in colon length, as well as a marked increase in monocyte/macrophage inflammatory infiltrates and histopathology changes in the colon. This was accompanied by a clear, inverse relationship between IFN-gamma and IL-10 responses in Slc11a1 wt compared with mt mice, resulting in a significantly higher ratio of IFN-gamma:IL-10 in wt compared with mt mice in lymph node and splenic T cells. RNase protection assays confirmed the presence of significantly higher IFN-gamma at the RNA level in the colons of wt compared with mt mice at Day 7 of treatment. Interestingly this was accompanied by significantly enhanced RNA levels for the acute-phase protein IL-6, which is known to inhibit the generation of forkhead box P3+ regulatory T cells and help to drive the differentiation of Th17 from naive T cells and not by differences in RNA for IL-12p35 or IL-12p40 molecules that dimerize to form the Th1-inducing cytokine IL-12.


Subject(s)
Cation Transport Proteins/physiology , Colitis/immunology , Dextran Sulfate/pharmacology , Animals , Colitis/chemically induced , Interferon-gamma/analysis , Interleukin-10/analysis , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p40/genetics , Interleukin-6/genetics , Lymph Nodes , Mice , RNA, Messenger/analysis , Spleen
4.
J Biol Chem ; 282(49): 35646-56, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17932044

ABSTRACT

Slc11a1 (formerly Nramp1) is a proton/divalent cation transporter that regulates cation homeostasis in macrophages. Slc11a2 mediates divalent cation uptake via the gut and delivery into cells. The mode of action of the two transporters remains controversial. Heterologous expression in frog oocytes shows Slc11a2 is a symporter, whereas Slc11a1 is an antiporter fluxing divalent cations against the proton gradient. This explains why Slc11a2, but not Slc11a1, can complement EGTA sensitivity in smf1Delta/smf2Delta/smf3Delta yeast. However, some studies of transport in mammalian cells suggest Slc11a1 is a symporter. We now demonstrate that Slc11a1, but not Slc11a2, complements a divalent cation stress phenotype in bsd2Delta/rer1Delta yeast. This is the first description of a yeast complementation assay for Slc11a1 function. Given the prior demonstration in frog oocytes that Slc11a1 acts as an antiporter, the most plausible interpretation of the data is that Slc11a1 is rescuing bsd2Delta/rer1Delta yeast by exporting divalent cations. Chimaeras define the N terminus, and a segment of the protein core preceding transmembrane domain 9 through transmembrane domain 12, as important in rescuing the divalent cation stress phenotype. EGTA sensitivity and divalent cation stress phenotypes in yeast expressing Slc11a orthologues show that symport activity is ancestral. Molecular changes that mediate rescue of the divalent cation stress phenotype post-date frogs and co-evolved with Slc11a1 orthologues that regulate divalent cation homeostasis in macrophages and resistance to infection in chickens and mammals.


Subject(s)
Cation Transport Proteins/metabolism , Evolution, Molecular , Mutant Chimeric Proteins/metabolism , Animals , Cation Transport Proteins/genetics , Cations, Divalent/metabolism , Chickens , Female , Gene Expression , Genetic Complementation Test , Humans , Ion Transport/genetics , Macrophages/metabolism , Mutant Chimeric Proteins/genetics , Oocytes/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Xenopus laevis
5.
Infect Immun ; 75(10): 5059-67, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17620357

ABSTRACT

Solute carrier family 11 member a1 (Slc11a1; formerly Nramp1) encodes a late endosomal/lysosomal protein/divalent cation transporter that regulates iron homeostasis in macrophages. During macrophage activation, Slc11a1 has multiple pleiotropic effects on gene regulation and function, including gamma interferon-induced class II expression and antigen-presenting cell function. The wild-type allele at Slc11a1 has been associated with a bias in Th1 cell function in vivo, which is beneficial in resistance to infection against intracellular macrophage pathogens but detrimental in contributing to development of type 1 diabetes. The extent to which this depends on macrophage versus dendritic cell (DC) function is not known. Here we show that Slc11a1 is expressed in late endosomes and/or lysosomes of CD11c(+) DCs. DCs from mutant and congenic wild-type mice upregulate interleukin-12 (IL-12) and IL-10 mRNA in response to lipopolysaccharide (LPS) stimulation, but the ratio of IL-10 to IL-12 is higher in unstimulated DCs and DCs stimulated for 15 h with LPS from mutant mice than from wild-type mice. DCs from wild-type mice upregulate major histocompatibility complex class II in response to LPS more efficiently than DCs from mutant mice. Unstimulated DCs from wild-type and mutant mice present ovalbumin (OVA) peptide with an efficiency equivalent to that of an OVA-specific CD4 T-cell line, but DCs from wild-type mice are more efficient at processing and presenting OVA or Leishmania activator of cell kinase (LACK) protein to OVA- and LACK-specific T cells. These data indicate that wild-type Slc11a1 expressed in DCs may play a role both in determining resistance to infectious disease and in susceptibility to autoimmune disease such as type 1 diabetes.


Subject(s)
Antigen Presentation/immunology , Cation Transport Proteins/biosynthesis , Dendritic Cells/immunology , Gene Expression Regulation , Histocompatibility Antigens Class II/biosynthesis , Animals , Antigens, Protozoan/immunology , Bias , Cation Transport Proteins/immunology , Cells, Cultured , Dendritic Cells/chemistry , Endosomes/chemistry , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-12/biosynthesis , Interleukin-12/genetics , Leishmania , Lipopolysaccharides/immunology , Lysosomes/chemistry , Major Histocompatibility Complex , Mice , Mice, Mutant Strains , Microscopy, Confocal , Ovalbumin/immunology , Protozoan Proteins/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
J Leukoc Biol ; 77(3): 311-20, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15601666

ABSTRACT

Solute carrier family 11a member 1 (Slc11a1; formerly natural resistance-associated macrophage protein 1) encodes a late endosomal/lysosomal protein/divalent cation transporter, which regulates iron homeostasis in macrophages. During macrophage activation, Slc11a1 exerts pleiotropic effects on gene regulation and function, including generation of nitric oxide (NO) via inducible NO synthase (iNOS; encoded by Nos2A) and of reactive oxygen intermediates (ROI) via the phagocyte oxidase complex. As NO and ROI have potent antimicrobial activity in macrophages, it was assumed that their activities would contribute to Slc11a1-regulated innate resistance to Salmonella enterica serovar Typhimurium and Leishmania donovani. By intercrossing mice with gene disruptions at Nos2A and Cybb (encoding gp91phox, the heavy chain subunit of cytochrome b-245 and an essential component of phagocyte NADPH oxidase) onto equivalent Slc11a1 wild-type and mutant genetic backgrounds, we demonstrate that neither iNOS nor gp91phox activity is required for Slc11a1-mediated innate resistance to either infection. Functional gp91phox and iNOS are required to control S. enterica serovar Typhimurium in non-Slc11a1-regulated phases of infection. For L. donovani, an organ-specific requirement for iNOS to clear parasites from the spleen was observed at 50 days post-infection, but neither iNOS nor gp91phox influenced late-phase infection in the liver. This contrasted with Leishmania major infection, which caused rapid lesion growth and death in iNOS knockout mice and some exacerbation of disease with gp91phox deficiency. This highlights the adaptive differences in tissue and cellular tropisms between L. donovani and L. major and the different genes and mechanisms that regulate visceral versus cutaneous forms of the disease.


Subject(s)
Cation Transport Proteins/metabolism , Leishmaniasis, Visceral/enzymology , NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Phagocytes/enzymology , Salmonella Infections, Animal/enzymology , Animals , Cation Transport Proteins/genetics , Disease Models, Animal , Genotype , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunohistochemistry , In Vitro Techniques , Leishmania donovani/pathogenicity , Leishmania major/pathogenicity , Leishmaniasis, Visceral/microbiology , Liver/microbiology , Liver/parasitology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Spleen/microbiology , Spleen/parasitology
7.
Biochem J ; 382(Pt 3): 811-9, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15202932

ABSTRACT

Solute carrier family 11 member 1 (Slc11a1, formerly Nramp1) is a highly glycosylated, 12 transmembrane domain protein expressed in macrophages. It resides in the membrane of late endosomes and lysosomes, where it functions as a bivalent cation transporter. Mice susceptible to infection by various intracellular pathogens including Leishmania donovani and Salmonella typhimurium carry a glycine to aspartic acid substitution at position 169 (G169D, Gly(169)-->Asp), within transmembrane domain 4 of Slc11a1. To investigate the molecular pathogenesis of infectious disease susceptibility, we compared the behaviour of heterologously and endogenously expressed wild-type and mutant Slc11a1 by immunofluorescence, immunoelectron microscopy and Western-blot analysis. We found occasional late endosome/lysosome staining of mutant protein using immunoelectron microscopy, but most of the mutant Slc11a1 was retained within the ER (endoplasmic reticulum). Using glycosylation as a marker for protein maturation in two independent heterologous expression systems, we found that most mutant Slc11a1 existed as an ER-dependent, partially glycosylated intermediate species. Correct endosomal targeting of wild-type Slc11a1 continued despite disruption of N-glycosylation sites, indicating that glycosylation did not influence folding or sorting. We propose that the G169D mutation causes localized misfolding of Slc11a1, resulting in its retention in the ER and manifestation of the loss of function phenotype.


Subject(s)
Cation Transport Proteins/metabolism , Organelles/metabolism , Animals , COS Cells , Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Cell Line , Chlorocebus aethiops , Communicable Diseases/genetics , Disease Susceptibility , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Glycosylation , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...