Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Extracell Vesicles ; 12(12): e12380, 2023 12.
Article in English | MEDLINE | ID: mdl-38010190

ABSTRACT

Extracellular vesicles (EVs) are lipid bilayer nanoparticles involved in cell-cell communication that are released into the extracellular space by all cell types. The cargo of EVs includes proteins, lipids, nucleic acids, and metabolites reflecting their cell of origin. EVs have recently been isolated directly from solid tissues, and this may provide insights into how EVs mediate communication between cells in vivo. Even though EVs have been isolated from tissues, their point of origin when they are in the interstitial space has been uncertain. In this study, we performed three-dimensional (3D) reconstruction using transmission electron tomography of metastatic and normal liver tissues with a focus on the presence of EVs in the interstitium. After chemical fixation of the samples and subsequent embedding of tissue pieces in resin, ultrathin slices (300 nm) were cut and imaged on a 120 ekV transmission electron microscopy as a tilt series (a series of subsequent images tilted at different angles). These were then computationally illustrated in a 3D manner to reconstruct the imaged tissue volume. We identified the cells delimiting the interstitial space in both types of tissues, and small distinct spherical structures with a diameter of 30-200 nm were identified between the cells. These round structures appeared to be more abundant in metastatic tissue compared to normal tissue. We suggest that the observed spherical structures in the interstitium of the metastatic and non-metastatic liver represent EVs. This work thus provides the first 3D visualization of EVs in human tissue.


Subject(s)
Extracellular Vesicles , Humans , Extracellular Vesicles/metabolism , Electron Microscope Tomography , Imaging, Three-Dimensional , Liver/diagnostic imaging , Microscopy, Electron, Transmission
4.
Elife ; 62017 05 02.
Article in English | MEDLINE | ID: mdl-28463107

ABSTRACT

cAMP/PKA signalling is compartmentalised with tight spatial and temporal control of signal propagation underpinning specificity of response. The cAMP-degrading enzymes, phosphodiesterases (PDEs), localise to specific subcellular domains within which they control local cAMP levels and are key regulators of signal compartmentalisation. Several components of the cAMP/PKA cascade are located to different mitochondrial sub-compartments, suggesting the presence of multiple cAMP/PKA signalling domains within the organelle. The function and regulation of these domains remain largely unknown. Here, we describe a novel cAMP/PKA signalling domain localised at mitochondrial membranes and regulated by PDE2A2. Using pharmacological and genetic approaches combined with real-time FRET imaging and high resolution microscopy, we demonstrate that in rat cardiac myocytes and other cell types mitochondrial PDE2A2 regulates local cAMP levels and PKA-dependent phosphorylation of Drp1. We further demonstrate that inhibition of PDE2A, by enhancing the hormone-dependent cAMP response locally, affects mitochondria dynamics and protects from apoptotic cell death.


Subject(s)
Apoptosis , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dynamins/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Animals , Cell Line , Humans , Mice , Phosphorylation , Protein Processing, Post-Translational , Rats
5.
Tissue Cell ; 49(2 Pt A): 131-132, 2017 04.
Article in English | MEDLINE | ID: mdl-28342448

Subject(s)
Golgi Apparatus , Animals , Humans
6.
J Hazard Mater ; 324(Pt A): 3-14, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-26952084

ABSTRACT

A putative biosynthetic mechanism for selenium nanoparticles (SeNPs) and efficient reduction of selenite (SeO32-) in the bacterial strain Stenotrophomonas maltophilia SeITE02 are addressed here on the basis of information gained by a combined approach relying on a set of physiological, chemical/biochemical, microscopy, and proteomic analyses. S. maltophilia SeITE02 is demonstrated to efficiently transform selenite into elemental selenium (Se°) by reducing 100% of 0.5mM of this toxic oxyanion to Se° nanoparticles within 48h growth, in liquid medium. Since the selenite reducing activity was detected in the cytoplasmic protein fraction, while biogenic SeNPs showed mainly extracellular localization, a releasing mechanism of SeNPs from the intracellular environment is hypothesized. SeNPs appeared spherical in shape and with size ranging from 160nm to 250nm, depending on the age of the cultures. Proteomic analysis carried out on the cytoplasmic fraction identified an alcohol dehydrogenase homolog, conceivably correlated with the biogenesis of SeNPs. Finally, by Fourier Transformed Infrared Spectrometry, protein and lipid residues were detected on the surface of biogenic SeNPs. Eventually, this strain might be efficaciously exploited for the remediation of selenite-contaminated environmental matrices due to its high SeO32- reducing efficiency. Biogenic SeNPs may also be considered for technological applications in different fields.


Subject(s)
Selenious Acid/chemistry , Stenotrophomonas maltophilia/metabolism , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Biodegradation, Environmental , Catalysis , Nanoparticles/metabolism , Oxidation-Reduction , Particle Size , Proteomics , Stenotrophomonas maltophilia/genetics
8.
J Immunol ; 195(8): 3890-900, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26371254

ABSTRACT

Cytokines are key regulators of adequate immune responses to infection with Mycobacterium tuberculosis. We demonstrate that the p110δ catalytic subunit of PI3K acts as a downstream effector of the TLR family member RP105 (CD180) in promoting mycobacteria-induced cytokine production by macrophages. Our data show that the significantly reduced release of TNF and IL-6 by RP105(-/-) macrophages during mycobacterial infection was not accompanied by diminished mRNA or protein expression. Mycobacteria induced comparable activation of NF-κB and p38 MAPK signaling in wild-type (WT) and RP105(-/-) macrophages. In contrast, mycobacteria-induced phosphorylation of Akt was abrogated in RP105(-/-) macrophages. The p110δ-specific inhibitor, Cal-101, and small interfering RNA-mediated knockdown of p110δ diminished mycobacteria-induced TNF secretion by WT but not RP105(-/-) macrophages. Such interference with p110δ activity led to reduced surface-expressed TNF in WT but not RP105(-/-) macrophages, while leaving TNF mRNA and protein expression unaffected. Activity of Bruton's tyrosine kinase was required for RP105-mediated activation of Akt phosphorylation and TNF release by mycobacteria-infected macrophages. These data unveil a novel innate immune signaling axis that orchestrates key cytokine responses of macrophages and provide molecular insight into the functions of RP105 as an innate immune receptor for mycobacteria.


Subject(s)
Antigens, CD/immunology , Class I Phosphatidylinositol 3-Kinases/immunology , MAP Kinase Signaling System/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens, CD/genetics , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Enzyme Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Protein Transport/drug effects , Protein Transport/genetics , Protein Transport/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Purines/pharmacology , Quinazolinones/pharmacology , Tuberculosis/genetics , Tuberculosis/pathology , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
9.
Appl Environ Microbiol ; 80(20): 6480-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25107981

ABSTRACT

The diversity of bacterial species in the human oral cavity is well recognized, but a high proportion of them are presently uncultivable. Candidate division TM7 bacteria are almost always detected in metagenomic studies but have not yet been cultivated. In this paper, we identified candidate division TM7 bacterial phylotypes in mature plaque samples from around orthodontic bonds in subjects undergoing orthodontic treatment. Successive rounds of enrichment in laboratory media led to the isolation of a pure culture of one of these candidate division TM7 phylotypes. The bacteria formed filaments of 20 to 200 µm in length within agar plate colonies and in monospecies biofilms on salivary pellicle and exhibited some unusual morphological characteristics by transmission electron microscopy, including a trilaminated cell surface layer and dense cytoplasmic deposits. Proteomic analyses of cell wall protein extracts identified abundant polypeptides predicted from the TM7 partial genomic sequence. Pleiomorphic phenotypes were observed when the candidate division TM7 bacterium was grown in dual-species biofilms with representatives of six different oral bacterial genera. The TM7 bacterium formed long filaments in dual-species biofilm communities with Actinomyces oris or Fusobacterium nucleatum. However, the TM7 isolate grew as short rods or cocci in dual-species biofilms with Porphyromonas gingivalis, Prevotella intermedia, Parvimonas micra, or Streptococcus gordonii, forming notably robust biofilms with the latter two species. The ability to cultivate TM7 axenically should majorly advance understanding of the physiology, genetics, and virulence properties of this novel candidate division oral bacterium.


Subject(s)
Axenic Culture , Bacteria/cytology , Bacteria/genetics , Mouth/microbiology , Actinomyces/growth & development , Actinomyces/physiology , Adolescent , Bacteria/classification , Bacteria/isolation & purification , Biofilms/growth & development , Child , Denaturing Gradient Gel Electrophoresis , Fusobacterium nucleatum/growth & development , Fusobacterium nucleatum/physiology , Humans , Molecular Sequence Data , Orthodontic Appliances/microbiology , Phylogeny , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/physiology , Proteomics/methods , RNA, Ribosomal, 16S , Streptococcus gordonii/growth & development , Streptococcus gordonii/physiology
10.
Immunol Cell Biol ; 92(3): 275-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24343664

ABSTRACT

Rho GTPases are required for many cellular events such as adhesion, motility, and membrane trafficking. Here we show that in macrophages, the Rho GTPases Rac1 and Cdc42 are involved in lamellipodia and filopodia formation, respectively, and that both of these Rho GTPases are essential for the efficient surface delivery of tumor necrosis factor (TNF) to the plasma membrane following TLR4 stimulation. We have previously demonstrated intracellular trafficking of TNF via recycling endosomes in lipopolysaccharide (LPS)-activated macrophages. Here, we further define a specific role for Rac1 in intracellular TNF trafficking, demonstrating impairment in TNF release following TLR4 stimulation in the presence of a Rac inhibitor, in cells expressing a dominant negative (DN) form of Rac1, and following small interfering RNA (siRNA) knockdown of Rac1. Rac1 activity was required for TNF trafficking but not for TLR4 signaling following LPS stimulation. Reduced TNF secretion was due to a defect in Rac1 activity, but not of the closely related Rho GTPase Rac2, demonstrated by the additional use of macrophages derived from Rac2-deficient mice. Labeling recycling endosomes by the uptake of fluorescent transferrin enabled us to show that Rac1 was required for the final stages of TNF trafficking and delivery from recycling endosomes to the plasma membrane. Thus, actin remodeling by the Rho GTPase Rac1 is required for TNF cell surface delivery and release from macrophages.


Subject(s)
Endocytosis , Endosomes/metabolism , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Gene Deletion , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Protein Transport/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , Pyrones/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors , RAC2 GTP-Binding Protein
11.
Traffic ; 14(5): 568-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23387339

ABSTRACT

The Golgi apparatus is the main glycosylation and sorting station along the secretory pathway. Its structure includes the Golgi vesicles, which are depleted of anterograde cargo, and also of at least some Golgi-resident proteins. The role of Golgi vesicles remains unclear. Here, we show that Golgi vesicles are enriched in the Qb-SNAREs GS27 (membrin) and GS28 (GOS-28), and depleted of nucleotide sugar transporters. A block of intra-Golgi transport leads to accumulation of Golgi vesicles and partitioning of GS27 and GS28 into these vesicles. Conversely, active intra-Golgi transport induces fusion of these vesicles with the Golgi cisternae, delivering GS27 and GS28 to these cisternae. In an in vitro assay based on a donor compartment that lacks UDP-galactose translocase (a sugar transporter), the segregation of Golgi vesicles from isolated Golgi membranes inhibits intra-Golgi transport; re-addition of isolated Golgi vesicles devoid of UDP-galactose translocase obtained from normal cells restores intra-Golgi transport. We conclude that this activity is due to the presence of GS27 and GS28 in the Golgi vesicles, rather than the sugar transporter. Furthermore, there is an inverse correlation between the number of Golgi vesicles and the number of inter-cisternal connections under different experimental conditions. Finally, a rapid block of the formation of vesicles via COPI through degradation of ϵCOP accelerates the cis-to-trans delivery of VSVG. These data suggest that Golgi vesicles, presumably with COPI, serve to inhibit intra-Golgi transport by the extraction of GS27 and GS28 from the Golgi cisternae, which blocks the formation of inter-cisternal connections.


Subject(s)
Golgi Apparatus/metabolism , Qb-SNARE Proteins/metabolism , Animals , Biological Transport , CHO Cells , Cricetinae , Cricetulus , Fibroblasts/metabolism , HeLa Cells , Hep G2 Cells , Humans , Liver/metabolism , Membrane Glycoproteins/metabolism , Monosaccharide Transport Proteins/metabolism , Nucleotides/metabolism , Rats , Viral Envelope Proteins/metabolism
12.
PLoS One ; 8(2): e57034, 2013.
Article in English | MEDLINE | ID: mdl-23437303

ABSTRACT

Lipopolysaccharide (LPS)-activated macrophages secrete pro-inflammatory cytokines, including tumor necrosis factor (TNF) to elicit innate immune responses. Secretion of these cytokines is also a major contributing factor in chronic inflammatory disease. In previous studies we have begun to elucidate the pathways and molecules that mediate the intracellular trafficking and secretion of TNF. Rab6a and Rab6a' (collectively Rab6) are trans-Golgi-localized GTPases known for roles in maintaining Golgi structure and Golgi-associated trafficking. We found that induction of TNF secretion by LPS promoted the selective increase of Rab6 expression. Depletion of Rab6 (via siRNA and shRNA) resulted in reorganization of the Golgi ribbon into more compact structures that at the resolution of electron microcopy consisted of elongated Golgi stacks that likely arose from fusion of smaller Golgi elements. Concomitantly, the delivery of TNF to the cell surface and subsequent release into the media was reduced. Dominant negative mutants of Rab6 had similar effects in disrupting TNF secretion. In live cells, Rab6-GFP were localized on trans-Golgi network (TGN)-derived tubular carriers demarked by the golgin p230. Rab6 depletion and inactive mutants altered carrier egress and partially reduced p230 membrane association. Our results show that Rab6 acts on TNF trafficking at the level of TGN exit in tubular carriers and our findings suggest Rab6 may stabilize p230 on the tubules to facilitate TNF transport. Both Rab6 isoforms are needed in macrophages for Golgi stack organization and for the efficient post-Golgi transport of TNF. This work provides new insights into Rab6 function and into the role of the Golgi complex in cytokine secretion in inflammatory macrophages.


Subject(s)
Golgi Apparatus/metabolism , Macrophages/metabolism , Tumor Necrosis Factors/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Autoantigens/metabolism , Brefeldin A/pharmacology , Cell Line , Cell Membrane/metabolism , Golgi Matrix Proteins , Membrane Proteins/metabolism , Mice , Mutation , Protein Stability , Protein Transport/drug effects , RNA Interference , rab GTP-Binding Proteins/genetics
13.
Int J Mol Sci ; 14(1): 674-83, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23344038

ABSTRACT

The ATP2C1 gene encodes for the secretory pathway calcium (Ca2+)-ATPase pump (SPCA1), which localizes along the secretory pathway, mainly in the trans-Golgi. The loss of one ATP2C1 allele causes Hailey-Hailey disease in humans but not mice. Examining differences in genomic organization between mouse and human we speculate that the overlap between ATP2C1 and ASTE1 genes only in humans could explain this different response to ATP2C1 dysregulation. We propose that ASTE1, overlapping with ATP2C1 in humans, affects alternative splicing, and potentially protein expression of the latter. If dysregulated, the composition of the SPCA1 isoform pool could diverge from the physiological status, affecting cytosolic Ca2+-signaling, and in turn perturbing cell division, leading to cell death or to neoplastic transformation.


Subject(s)
Calcium-Transporting ATPases/genetics , Gene Expression Regulation , Genes, Overlapping/genetics , Genome, Human/genetics , Proteins/genetics , Alternative Splicing , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Genetic Predisposition to Disease/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Pemphigus, Benign Familial/genetics , Pemphigus, Benign Familial/metabolism , Species Specificity
14.
PLoS One ; 7(6): e37352, 2012.
Article in English | MEDLINE | ID: mdl-22701565

ABSTRACT

BACKGROUND: Protein mono-ADP-ribosylation is a reversible post-translational modification that modulates the function of target proteins. The enzymes that catalyze this reaction in mammalian cells are either bacterial pathogenic toxins or endogenous cellular ADP-ribosyltransferases. The latter include members of three different families of proteins: the well characterized arginine-specific ecto-enzymes ARTCs, two sirtuins and, more recently, novel members of the poly(ADP-ribose) polymerase (PARP/ARTD) family that have been suggested to act as cellular mono-ADP-ribosyltransferases. Here, we report on the characterisation of human ARTD15, the only known ARTD family member with a putative C-terminal transmembrane domain. METHODOLOGY/PRINCIPAL FINDINGS: Immunofluorescence and electron microscopy were performed to characterise the sub-cellular localisation of ARTD15, which was found to be associated with membranes of the nuclear envelope and endoplasmic reticulum. The orientation of ARTD15 was determined using protease protection assay, and is shown to be a tail-anchored protein with a cytosolic catalytic domain. Importantly, by combining immunoprecipitation with mass spectrometry and using cell lysates from cells over-expressing FLAG-ARTD15, we have identified karyopherin-ß1, a component of the nuclear trafficking machinery, as a molecular partner of ARTD15. Finally, we demonstrate that ARTD15 is a mono-ADP-ribosyltransferase able to induce the ADP-ribosylation of karyopherin-ß1, thus defining the first substrate for this enzyme. CONCLUSIONS/SIGNIFICANCE: Our data reveal that ARTD15 is a novel ADP-ribosyltransferase enzyme with a new intracellular location. Finally, the identification of karyopherin-ß1 as a target of ARTD15-mediated ADP-ribosylation, hints at a novel regulatory mechanism of karyopherin-ß1 functions.


Subject(s)
ADP Ribose Transferases/metabolism , Endoplasmic Reticulum/enzymology , Nuclear Envelope/enzymology , Poly(ADP-ribose) Polymerases/metabolism , beta Karyopherins/metabolism , Endoplasmic Reticulum/ultrastructure , Fluorescent Antibody Technique , Humans , Mass Spectrometry , Microscopy, Electron , Nuclear Envelope/ultrastructure , Poly(ADP-ribose) Polymerases/genetics , Protein Structure, Tertiary
15.
Adv Exp Med Biol ; 740: 439-60, 2012.
Article in English | MEDLINE | ID: mdl-22453953

ABSTRACT

As with other complex cellular functions, intracellular membrane transport involves the coordinated engagement of a series of organelles and machineries; in the last couple of decades more importance has been given to the role of calcium (Ca(2+)) in the regulation of membrane trafficking, which is directly involved in coordinating the endoplasmic reticulum-to-Golgi-to-plasma membrane delivery of cargo. Consequently, the Golgi apparatus (GA) is now considered not just the place proteins mature in as they move to their final destination(s), but it is increasingly viewed as an intracellular Ca(2+) store. In the last few years the mechanisms regulating the homeostasis of Ca(2+) in the GA and its role in membrane trafficking have begun to be elucidated. Here, these recent discoveries that shed light on the role Ca(2+) plays as of trigger of different steps during membrane trafficking has been reviewed. This includes recruitment of proteins and SNARE cofactors to the Golgi membranes, which are both fundamental for the membrane remodeling and the regulation of fusion/fission events occurring during the passage of cargo across the GA. I conclude by focusing attention on Ca(2+) homeostasis dysfunctions in the GA and their related pathological implications.


Subject(s)
Calcium/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Animals , Biological Transport , Calcium-Transporting ATPases/physiology , Humans , Phospholipases A2/physiology , SNARE Proteins/physiology
16.
Traffic ; 13(5): 727-44, 2012 May.
Article in English | MEDLINE | ID: mdl-22335553

ABSTRACT

We have shown previously that Rab6, a small, trans-Golgi-localized GTPase, acts upstream of the conserved oligomeric Golgi complex (COG) and ZW10/RINT1 retrograde tether complexes to maintain Golgi homeostasis. In this article, we present evidence from the unbiased and high-resolution approach of electron microscopy and electron tomography that Rab6 is essential to the trans-Golgi trafficking of two morphological classes of coated vesicles; the larger corresponds to clathrin-coated vesicles and the smaller to coat protein I (COPI)-coated vesicles. On the basis of the site of coated vesicle accumulation, cisternal dilation and the normal kinetics of cargo transport from the endoplasmic reticulum (ER) to Golgi followed by delayed Golgi to cell surface transport, we suggest that Golgi function in cargo transport is preferentially inhibited at the trans-Golgi/trans-Golgi network (TGN). The >50% increase in Golgi cisternae number in Rab6-depleted HeLa cells that we observed may well be coupled to the trans-Golgi accumulation of COPI-coated vesicles; depletion of the individual Rab6 effector, myosin IIA, produced an accumulation of uncoated vesicles with if anything a decrease in cisternal number. These results are the first evidence for a Rab6-dependent protein machine affecting Golgi-proximal, coated vesicle accumulation and probably transport at the trans-Golgi and the first example of concomitant cisternal proliferation and increased Golgi stack organization under inhibited transport conditions.


Subject(s)
COP-Coated Vesicles/metabolism , Electron Microscope Tomography/methods , Golgi Apparatus/metabolism , rab GTP-Binding Proteins/metabolism , Biological Transport , Green Fluorescent Proteins/metabolism , HeLa Cells , Homeostasis , Humans , Image Processing, Computer-Assisted , Microscopy, Electron/methods , Myosin Type II/metabolism , Phenotype , Protein Isoforms , Protein Transport , RNA, Small Interfering/metabolism , trans-Golgi Network/metabolism
17.
Nature ; 477(7363): 225-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849978

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid cycle (TCA cycle) that catalyses the hydration of fumarate into malate. Germline mutations of FH are responsible for hereditary leiomyomatosis and renal-cell cancer (HLRCC). It has previously been demonstrated that the absence of FH leads to the accumulation of fumarate, which activates hypoxia-inducible factors (HIFs) at normal oxygen tensions. However, so far no mechanism that explains the ability of cells to survive without a functional TCA cycle has been provided. Here we use newly characterized genetically modified kidney mouse cells in which Fh1 has been deleted, and apply a newly developed computer model of the metabolism of these cells to predict and experimentally validate a linear metabolic pathway beginning with glutamine uptake and ending with bilirubin excretion from Fh1-deficient cells. This pathway, which involves the biosynthesis and degradation of haem, enables Fh1-deficient cells to use the accumulated TCA cycle metabolites and permits partial mitochondrial NADH production. We predicted and confirmed that targeting this pathway would render Fh1-deficient cells non-viable, while sparing wild-type Fh1-containing cells. This work goes beyond identifying a metabolic pathway that is induced in Fh1-deficient cells to demonstrate that inhibition of haem oxygenation is synthetically lethal when combined with Fh1 deficiency, providing a new potential target for treating HLRCC patients.


Subject(s)
Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genes, Lethal/genetics , Genes, Tumor Suppressor , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Mutation/genetics , Animals , Bilirubin/metabolism , Cell Line , Cells, Cultured , Citric Acid Cycle , Computer Simulation , Fumarate Hydratase/deficiency , Fumarates/metabolism , Glutamine/metabolism , Heme/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Leiomyomatosis/congenital , Leiomyomatosis/drug therapy , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Mice , Mitochondria/metabolism , NAD/metabolism , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms
18.
Traffic ; 11(10): 1315-33, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20604898

ABSTRACT

The Golgi apparatus (GA) is a dynamic store of Ca(2+) that can be released into the cell cytosol. It can thus participate in the regulation of the Ca(2+) concentration in the cytosol ([Ca(2+) ](cyt) ), which might be critical for intra-Golgi transport. Secretory pathway Ca(2+) -ATPase pump type 1 (SPCA1) is important in Golgi homeostasis of Ca(2+) . The subcellular localization of SPCA1 appears to be GA specific, although its precise location within the GA is not known. Here, we show that SPCA1 is mostly excluded from the cores of the Golgi cisternae and is instead located mainly on the lateral rims of Golgi stacks, in tubular noncompact zones that interconnect different Golgi stacks, and within tubular parts of the trans Golgi network, suggesting a role in regulation of the local [Ca(2+) ](cyt) that is crucial for membrane fusion. SPCA1 knockdown by RNA interference induces GA fragmentation. These Golgi fragments lack the cis-most and trans-most cisternae and remain within the perinuclear region. This SPCA1 knockdown inhibits exit of vesicular stomatitis virus G-protein from the GA and delays retrograde redistribution of the GA glycosylation enzymes into the endoplasmic reticulum caused by brefeldin A; however, exit of these enzymes from the endoplasmic reticulum is not affected. Thus, correct SPCA1 functioning is crucial to intra-Golgi transport and maintenance of the Golgi ribbon.


Subject(s)
Calcium-Transporting ATPases/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Brefeldin A/pharmacology , Calcium/metabolism , Calcium-Transporting ATPases/genetics , Cell Line , Cytosol/drug effects , Cytosol/metabolism , Cytosol/ultrastructure , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/ultrastructure , Protein Transport/drug effects
19.
Exp Cell Res ; 316(13): 2071-86, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20420828

ABSTRACT

The mechanisms of secretory transport through the Golgi apparatus remain an issue of debate. The precise functional importance of calcium ions (Ca(2+)) for intra-Golgi transport has also been poorly studied. Here, using different approaches to measure free Ca(2+) concentrations in the cell cytosol ([Ca(2+)](cyt)) and inside the lumen of the Golgi apparatus ([Ca(2+)](GA)), we have revealed transient increases in [Ca(2+)](cyt) during the late phase of intra-Golgi transport that are concomitant with a decline in the maximal [Ca(2+)](GA) restoration ability. Thus, this redistribution of Ca(2+) from the Golgi apparatus into the cytosol during the movement of cargo through the Golgi apparatus appears to have a role in intra-Golgi transport, and mainly in the late Ca(2+)-dependent phase of SNARE-regulated fusion of Golgi compartments.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Golgi Apparatus/metabolism , Biological Transport , Calcium Signaling , Cells, Cultured , Fibroblasts/metabolism , HeLa Cells , Humans , Skin/cytology , Skin/metabolism , Subcellular Fractions
20.
Proc Natl Acad Sci U S A ; 107(13): 5863-8, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231454

ABSTRACT

The size and integrity of the Golgi apparatus is maintained via a tightly controlled regulation of membrane traffic using a variety of different signaling and cytoskeletal proteins. We have recently observed that activation of c-Src has profound effects on Golgi structure, leading to dramatically vesiculated cisternae in a variety of cell types. As the large GTPase dynamin (Dyn2) has been implicated in Golgi vesiculation during secretion, we tested whether inhibiting Dyn2 activity by expression of a Dyn2K44A mutant or siRNA knockdown could attenuate active Src-induced Golgi fragmentation. Indeed, these perturbations attenuated fragmentation, and expression of a Dyn2Y(231/597)F mutant protein that cannot be phosphorylated by Src kinase had a similar effect . Finally, we find that Dyn2 is markedly phosphorylated during the transit of VSV-G protein through the TGN whereas expression of the Dyn2Y(231/597)F mutant significantly reduces exit of the nascent protein from this compartment. These findings demonstrate that activation of Dyn2 by Src kinase regulates Golgi integrity and vesiculation during the secretory process.


Subject(s)
Dynamin II/metabolism , Golgi Apparatus/metabolism , src-Family Kinases/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Cells, Cultured , Cricetinae , Dynamin II/antagonists & inhibitors , Dynamin II/genetics , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mutagenesis, Site-Directed , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenotype , Phosphorylation , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , trans-Golgi Network/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...