Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
Nature ; 606(7915): 761-768, 2022 06.
Article in English | MEDLINE | ID: mdl-35551511

ABSTRACT

SARS-CoV-2, like other coronaviruses, builds a membrane-bound replication organelle to enable RNA replication1. The SARS-CoV-2 replication organelle is composed of double-membrane vesicles (DMVs) that are tethered to the endoplasmic reticulum (ER) by thin membrane connectors2, but the viral proteins and the host factors involved remain unknown. Here we identify the viral non-structural proteins (NSPs) that generate the SARS-CoV-2 replication organelle. NSP3 and NSP4 generate the DMVs, whereas NSP6, through oligomerization and an amphipathic helix, zippers ER membranes and establishes the connectors. The NSP6(ΔSGF) mutant, which arose independently in the Alpha, Beta, Gamma, Eta, Iota and Lambda variants of SARS-CoV-2, behaves as a gain-of-function mutant with a higher ER-zippering activity. We identified three main roles for NSP6: first, to act as a filter in communication between the replication organelle and the ER, by allowing lipid flow but restricting the access of ER luminal proteins to the DMVs; second, to position and organize DMV clusters; and third, to mediate contact with lipid droplets (LDs) through the LD-tethering complex DFCP1-RAB18. NSP6 thus acts as an organizer of DMV clusters and can provide a selective means of refurbishing them with LD-derived lipids. Notably, both properly formed NSP6 connectors and LDs are required for the replication of SARS-CoV-2. Our findings provide insight into the biological activity of NSP6 of SARS-CoV-2 and of other coronaviruses, and have the potential to fuel the search for broad antiviral agents.


Subject(s)
Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Viral Nonstructural Proteins , Virus Replication , COVID-19/virology , Carrier Proteins , Cell Line , Coronavirus Nucleocapsid Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Humans , Lipid Droplets , SARS-CoV-2/genetics , SARS-CoV-2/growth & development , Viral Nonstructural Proteins/metabolism , rab GTP-Binding Proteins
2.
Traffic ; 25(1): e12924, 2024 01.
Article in English | MEDLINE | ID: mdl-37963679

ABSTRACT

The skeletal dysplasia spondyloepiphyseal dysplasia tarda (SEDT) is caused by mutations in the TRAPPC2 gene, which encodes Sedlin, a component of the trafficking protein particle (TRAPP) complex that we have shown previously to be required for the export of type II collagen (Col2) from the endoplasmic reticulum. No vertebrate model for SEDT has been generated thus far. To address this gap, we generated a Sedlin knockout animal by mutating the orthologous TRAPPC2 gene (olSedl) of Oryzias latipes (medaka) fish. OlSedl deficiency leads to embryonic defects, short size, diminished skeletal ossification and altered Col2 production and secretion, resembling human defects observed in SEDT patients. Moreover, SEDT knock-out animals display photoreceptor degeneration and gut morphogenesis defects, suggesting a key role for Sedlin in the development of these organs. Thus, by studying Sedlin function in vivo, we provide evidence for a mechanistic link between TRAPPC2-mediated membrane trafficking, Col2 export, and developmental disorders.


Subject(s)
Oryzias , Osteochondrodysplasias , Animals , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Oryzias/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Mutation , Osteochondrodysplasias/genetics
3.
Traffic ; 25(1): e12920, 2024 01.
Article in English | MEDLINE | ID: mdl-37886910

ABSTRACT

Wilson disease (WD) is caused by mutations in the ATP7B gene that encodes a copper (Cu) transporting ATPase whose trafficking from the Golgi to endo-lysosomal compartments drives sequestration of excess Cu and its further excretion from hepatocytes into the bile. Loss of ATP7B function leads to toxic Cu overload in the liver and subsequently in the brain, causing fatal hepatic and neurological abnormalities. The limitations of existing WD therapies call for the development of new therapeutic approaches, which require an amenable animal model system for screening and validation of drugs and molecular targets. To achieve this objective, we generated a mutant Caenorhabditis elegans strain with a substitution of a conserved histidine (H828Q) in the ATP7B ortholog cua-1 corresponding to the most common ATP7B variant (H1069Q) that causes WD. cua-1 mutant animals exhibited very poor resistance to Cu compared to the wild-type strain. This manifested in a strong delay in larval development, a shorter lifespan, impaired motility, oxidative stress pathway activation, and mitochondrial damage. In addition, morphological analysis revealed several neuronal abnormalities in cua-1 mutant animals exposed to Cu. Further investigation suggested that mutant CUA-1 is retained and degraded in the endoplasmic reticulum, similarly to human ATP7B-H1069Q. As a consequence, the mutant protein does not allow animals to counteract Cu toxicity. Notably, pharmacological correctors of ATP7B-H1069Q reduced Cu toxicity in cua-1 mutants indicating that similar pathogenic molecular pathways might be activated by the H/Q substitution and, therefore, targeted for rescue of ATP7B/CUA-1 function. Taken together, our findings suggest that the newly generated cua-1 mutant strain represents an excellent model for Cu toxicity studies in WD.


Subject(s)
Hepatolenticular Degeneration , Animals , Humans , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Copper/toxicity , Copper/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatocytes/metabolism
4.
Hum Mol Genet ; 32(7): 1090-1101, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36300303

ABSTRACT

Cysteamine is currently the only therapy for nephropathic cystinosis. It significantly improves life expectancy and delays progression to end-stage kidney disease; however, it cannot prevent it. Unfortunately, compliance to therapy is often weak, particularly during adolescence. Therefore, finding better treatments is a priority in the field of cystinosis. Previously, we found that genistein, an isoflavone particularly enriched in soy, can revert part of the cystinotic cellular phenotype that is not sensitive to cysteamine in vitro. To test the effects of genistein in vivo, we fed 2-month-old wild-type and Ctns-/- female mice with either a control diet, a genistein-containing diet or a cysteamine-containing diet for 14 months. Genistein (160 mg/kg/day) did not affect the growth of the mice or hepatic functionality. Compared with untreated mice at 16 months, Ctns-/- mice fed with genistein had lower cystine concentrations in their kidneys, reduced formation of cystine crystals, a smaller number of LAMP1-positive structures and an overall better-preserved parenchymal architecture. Cysteamine (400 mg/kg/day) was efficient in reverting the lysosomal phenotype and in preventing the development of renal lesions. These preclinical data indicate that genistein ameliorates kidney injury resulting from cystinosis with no side effects. Genistein therapy represents a potential treatment to improve the outcome for patients with cystinosis.


Subject(s)
Cystinosis , Kidney Diseases , Animals , Female , Mice , Cysteamine/therapeutic use , Cystine/therapeutic use , Cystinosis/drug therapy , Cystinosis/genetics , Disease Models, Animal , Genistein/pharmacology , Genistein/therapeutic use , Kidney
5.
EMBO J ; 39(8): e102468, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32154600

ABSTRACT

Vertebrate vision relies on the daily phagocytosis and lysosomal degradation of photoreceptor outer segments (POS) within the retinal pigment epithelium (RPE). However, how these events are controlled by light is largely unknown. Here, we show that the light-responsive miR-211 controls lysosomal biogenesis at the beginning of light-dark transitions in the RPE by targeting Ezrin, a cytoskeleton-associated protein essential for the regulation of calcium homeostasis. miR-211-mediated down-regulation of Ezrin leads to Ca2+ influx resulting in the activation of calcineurin, which in turn activates TFEB, the master regulator of lysosomal biogenesis. Light-mediated induction of lysosomal biogenesis and function is impaired in the RPE from miR-211-/- mice that show severely compromised vision. Pharmacological restoration of lysosomal biogenesis through Ezrin inhibition rescued the miR-211-/- phenotype, pointing to a new therapeutic target to counteract retinal degeneration associated with lysosomal dysfunction.


Subject(s)
Calcium/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , Lysosomes/metabolism , MicroRNAs/metabolism , Animals , Autophagy , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Down-Regulation , Light , Lysosomes/ultrastructure , Mice , Mice, Knockout , MicroRNAs/genetics , Phagocytosis , Phagosomes/metabolism , Phagosomes/ultrastructure , Retinal Pigment Epithelium/metabolism
6.
EMBO J ; 39(17): e105696, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32716134

ABSTRACT

Lysosomal degradation of the endoplasmic reticulum (ER) via autophagy (ER-phagy) is emerging as a critical regulator of cell homeostasis and function. The recent identification of ER-phagy receptors has shed light on the molecular mechanisms underlining this process. However, the signaling pathways regulating ER-phagy in response to cellular needs are still largely unknown. We found that the nutrient responsive transcription factors TFEB and TFE3-master regulators of lysosomal biogenesis and autophagy-control ER-phagy by inducing the expression of the ER-phagy receptor FAM134B. The TFEB/TFE3-FAM134B axis promotes ER-phagy activation upon prolonged starvation. In addition, this pathway is activated in chondrocytes by FGF signaling, a critical regulator of skeletal growth. FGF signaling induces JNK-dependent proteasomal degradation of the insulin receptor substrate 1 (IRS1), which in turn inhibits the PI3K-PKB/Akt-mTORC1 pathway and promotes TFEB/TFE3 nuclear translocation and enhances FAM134B transcription. Notably, FAM134B is required for protein secretion in chondrocytes, and cartilage growth and bone mineralization in medaka fish. This study identifies a new signaling pathway that allows ER-phagy to respond to both metabolic and developmental cues.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Nucleus/genetics , Endoplasmic Reticulum/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/genetics , Mice , Oryzias
7.
Mol Ther ; 31(9): 2651-2661, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37394797

ABSTRACT

Mutant Z alpha-1 antitrypsin (ATZ) accumulates in globules in the liver and is the prototype of proteotoxic hepatic disease. Therapeutic strategies aiming at clearance of polymeric ATZ are needed. Transient receptor potential mucolipin-1 (TRPML1) is a lysosomal Ca2+ channel that maintains lysosomal homeostasis. In this study, we show that by increasing lysosomal exocytosis, TRPML1 gene transfer or small-molecule-mediated activation of TRPML1 reduces hepatic ATZ globules and fibrosis in PiZ transgenic mice that express the human ATZ. ATZ globule clearance induced by TRPML1 occurred without increase in autophagy or nuclear translocation of TFEB. Our results show that targeting TRPML1 and lysosomal exocytosis is a novel approach for treatment of the liver disease due to ATZ and potentially other diseases due to proteotoxic liver storage.


Subject(s)
Liver Diseases , Transient Receptor Potential Channels , alpha 1-Antitrypsin , Animals , Humans , Mice , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , Liver Diseases/metabolism , Lysosomes/metabolism , Mice, Transgenic , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
8.
Orthod Craniofac Res ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38800926

ABSTRACT

INTRODUCTION: The aim of this in vitro study was to examine the potential impact of different curing times of 3D-printed orthodontic aligners on their cytotoxicity. METHOD: Some 60 samples of aligner material were directly 3D printed using Tera Harz TC-85 DAC resin and randomly allocated to three different curing time groups (14, 24 and 50 min). Zendura FLX samples were used as control. The samples were incubated in saliva for 14 days, and then the supernatant was collected. Human gingival fibroblasts (HGF-1)-CRL2014 were used to evaluate potential cytotoxicity. Furthermore, HGF-1 cells were plated on the samples as well as on a glass control sample. After 72 h of growth, their viability was tested. RESULTS: Compared with the glass, only the 50-min curing time markedly reduced fibroblast cell growth. Additionally, a negative linear trend was observed between curing time and fibroblast growth. In comparison with the aligner control group, all samples, including the aligner control samples, exhibited a significant reduction in the viability of human fibroblasts when exposed to saliva. CONCLUSIONS: 3D directly printed aligners showed a cytotoxic effect similar to that of thermoformed conventional aligners in terms of fibroblasts growth. A linear trend was found between curing time and cells growth, indicating that directly printed aligners could exhibit higher cytotoxicity if exposed to a longer curing time. This dependence on curing time underscores the importance of following a strict manufacturing process.

9.
EMBO J ; 38(19): e101704, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31429971

ABSTRACT

The TRAnsport Protein Particle (TRAPP) complex controls multiple membrane trafficking steps and is strategically positioned to mediate cell adaptation to diverse environmental conditions, including acute stress. We have identified the TRAPP complex as a component of a branch of the integrated stress response that impinges on the early secretory pathway. The TRAPP complex associates with and drives the recruitment of the COPII coat to stress granules (SGs) leading to vesiculation of the Golgi complex and arrest of ER export. The relocation of the TRAPP complex and COPII to SGs only occurs in cycling cells and is CDK1/2-dependent, being driven by the interaction of TRAPP with hnRNPK, a CDK substrate that associates with SGs when phosphorylated. In addition, CDK1/2 inhibition impairs TRAPP complex/COPII relocation to SGs while stabilizing them at ER exit sites. Importantly, the TRAPP complex controls the maturation of SGs. SGs that assemble in TRAPP-depleted cells are smaller and are no longer able to recruit RACK1 and Raptor, two TRAPP-interactive signaling proteins, sensitizing cells to stress-induced apoptosis.


Subject(s)
COP-Coated Vesicles/metabolism , Membrane Transport Proteins/metabolism , Stress, Physiological , Animals , CDC2 Protein Kinase/metabolism , Cell Line , Cyclin-Dependent Kinase 2/metabolism , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Rats
10.
EMBO Rep ; 22(9): e52289, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34338405

ABSTRACT

Degradation of the endoplasmic reticulum (ER) via selective autophagy (ER-phagy) is vital for cellular homeostasis. We identify FAM134A/RETREG2 and FAM134C/RETREG3 as ER-phagy receptors, which predominantly exist in an inactive state under basal conditions. Upon autophagy induction and ER stress signal, they can induce significant ER fragmentation and subsequent lysosomal degradation. FAM134A, FAM134B/RETREG1, and FAM134C are essential for maintaining ER morphology in a LC3-interacting region (LIR)-dependent manner. Overexpression of any FAM134 paralogue has the capacity to significantly augment the general ER-phagy flux upon starvation or ER-stress. Global proteomic analysis of FAM134 overexpressing and knockout cell lines reveals several protein clusters that are distinctly regulated by each of the FAM134 paralogues as well as a cluster of commonly regulated ER-resident proteins. Utilizing pro-Collagen I, as a shared ER-phagy substrate, we observe that FAM134A acts in a LIR-independent manner and compensates for the loss of FAM134B and FAM134C, respectively. FAM134C instead is unable to compensate for the loss of its paralogues. Taken together, our data show that FAM134 paralogues contribute to common and unique ER-phagy pathways.


Subject(s)
Membrane Proteins , Proteomics , Autophagy/genetics , Collagen , Endoplasmic Reticulum/genetics , Membrane Proteins/genetics , Quality Control
11.
Mol Ther ; 30(4): 1432-1450, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35121108

ABSTRACT

Mucopolysaccharidosis type IIIA (MPS-IIIA) is an autosomal recessive disorder caused by mutations in SGSH involved in the degradation of heparan sulfate. MPS-IIIA presents severe neurological symptoms such as progressive developmental delay and cognitive decline, for which there is currently no treatment. Brain targeting represents the main challenge for therapeutics to treat MPS-IIIA, and the development of small-molecule-based treatments able to reach the CNS could be a relevant advance for therapy. Using cell-based high content imaging to survey clinically approved drugs in MPS-IIIA cells, we identified fluoxetine, a selective serotonin reuptake inhibitor. Fluoxetine increases lysosomal and autophagic functions via TFEB activation through a RagC-dependent mechanism. Mechanistically, fluoxetine increases lysosomal exocytosis in mouse embryonic fibroblasts from MPS-IIIA mice, suggesting that this process may be responsible for heparan sulfate clearance. In vivo, fluoxetine ameliorates somatic and brain pathology in a mouse model of MPS-IIIA by decreasing the accumulation of glycosaminoglycans and aggregated autophagic substrates, reducing inflammation, and slowing down cognitive deterioration. We repurposed fluoxetine for potential therapeutics to treat human MPS-IIIA disease.


Subject(s)
Mucopolysaccharidosis III , Animals , Disease Models, Animal , Fibroblasts/metabolism , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Heparitin Sulfate/metabolism , Hydrolases/genetics , Mice , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/genetics
12.
Nature ; 528(7581): 272-5, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26595272

ABSTRACT

Skeletal growth relies on both biosynthetic and catabolic processes. While the role of the former is clearly established, how the latter contributes to growth-promoting pathways is less understood. Macroautophagy, hereafter referred to as autophagy, is a catabolic process that plays a fundamental part in tissue homeostasis. We investigated the role of autophagy during bone growth, which is mediated by chondrocyte rate of proliferation, hypertrophic differentiation and extracellular matrix (ECM) deposition in growth plates. Here we show that autophagy is induced in growth-plate chondrocytes during post-natal development and regulates the secretion of type II collagen (Col2), the major component of cartilage ECM. Mice lacking the autophagy related gene 7 (Atg7) in chondrocytes experience endoplasmic reticulum storage of type II procollagen (PC2) and defective formation of the Col2 fibrillary network in the ECM. Surprisingly, post-natal induction of chondrocyte autophagy is mediated by the growth factor FGF18 through FGFR4 and JNK-dependent activation of the autophagy initiation complex VPS34-beclin-1. Autophagy is completely suppressed in growth plates from Fgf18(-/-) embryos, while Fgf18(+/-) heterozygous and Fgfr4(-/-) mice fail to induce autophagy during post-natal development and show decreased Col2 levels in the growth plate. Strikingly, the Fgf18(+/-) and Fgfr4(-/-) phenotypes can be rescued in vivo by pharmacological activation of autophagy, pointing to autophagy as a novel effector of FGF signalling in bone. These data demonstrate that autophagy is a developmentally regulated process necessary for bone growth, and identify FGF signalling as a crucial regulator of autophagy in chondrocytes.


Subject(s)
Autophagy/physiology , Bone Development/physiology , Fibroblast Growth Factors/genetics , Signal Transduction , Animals , Autophagy/genetics , Autophagy-Related Protein 7 , Bone Development/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type II/metabolism , Embryo, Mammalian , Extracellular Matrix/genetics , Fibroblast Growth Factors/metabolism , Growth Plate/cytology , Growth Plate/metabolism , MAP Kinase Signaling System , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism
13.
Nature ; 525(7568): 234-8, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26354482

ABSTRACT

The amount of ice present in clouds can affect cloud lifetime, precipitation and radiative properties. The formation of ice in clouds is facilitated by the presence of airborne ice-nucleating particles. Sea spray is one of the major global sources of atmospheric particles, but it is unclear to what extent these particles are capable of nucleating ice. Sea-spray aerosol contains large amounts of organic material that is ejected into the atmosphere during bubble bursting at the organically enriched sea-air interface or sea surface microlayer. Here we show that organic material in the sea surface microlayer nucleates ice under conditions relevant for mixed-phase cloud and high-altitude ice cloud formation. The ice-nucleating material is probably biogenic and less than approximately 0.2 micrometres in size. We find that exudates separated from cells of the marine diatom Thalassiosira pseudonana nucleate ice, and propose that organic material associated with phytoplankton cell exudates is a likely candidate for the observed ice-nucleating ability of the microlayer samples. Global model simulations of marine organic aerosol, in combination with our measurements, suggest that marine organic material may be an important source of ice-nucleating particles in remote marine environments such as the Southern Ocean, North Pacific Ocean and North Atlantic Ocean.


Subject(s)
Atmosphere/chemistry , Ice , Aerosols/chemical synthesis , Aerosols/chemistry , Air , Aquatic Organisms/chemistry , Arctic Regions , Diatoms/chemistry , Freezing , Organic Chemicals/analysis , Organic Chemicals/chemistry , Phytoplankton/chemistry , Seawater/chemistry
14.
Proc Natl Acad Sci U S A ; 115(2): 391-396, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29279371

ABSTRACT

Ammonia is a potent neurotoxin that is detoxified mainly by the urea cycle in the liver. Hyperammonemia is a common complication of a wide variety of both inherited and acquired liver diseases. If not treated early and thoroughly, it results in encephalopathy and death. Here, we found that hepatic autophagy is critically involved in systemic ammonia homeostasis by providing key urea-cycle intermediates and ATP. Hepatic autophagy is triggered in vivo by hyperammonemia through an α-ketoglutarate-dependent inhibition of the mammalian target of rapamycin complex 1, and deficiency of autophagy impairs ammonia detoxification. In contrast, autophagy enhancement by means of hepatic gene transfer of the master regulator of autophagy transcription factor EB or treatments with the autophagy enhancers rapamycin and Tat-Beclin-1 increased ureagenesis and protected against hyperammonemia in a variety of acute and chronic hyperammonemia animal models, including acute liver failure and ornithine transcarbamylase deficiency, the most frequent urea-cycle disorder. In conclusion, hepatic autophagy is an important mechanism for ammonia detoxification because of its support of urea synthesis, and its enhancement has potential for therapy of both primary and secondary causes of hyperammonemia.


Subject(s)
Autophagy , Hyperammonemia/metabolism , Liver/metabolism , Urea/metabolism , Ammonia/metabolism , Animals , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
15.
Int J Mol Sci ; 22(11)2021 May 23.
Article in English | MEDLINE | ID: mdl-34071094

ABSTRACT

Three main approaches are used to combat severe viral respiratory infections. The first is preemptive vaccination that blocks infection. Weakened or dead viral particles, as well as genetic constructs carrying viral proteins or information about them, are used as an antigen. However, the viral genome is very evolutionary labile and changes continuously. Second, chemical agents are used during infection and inhibit the function of a number of viral proteins. However, these drugs lose their effectiveness because the virus can rapidly acquire resistance to them. The third is the search for points in the host metabolism the effect on which would suppress the replication of the virus but would not have a significant effect on the metabolism of the host. Here, we consider the possibility of using the copper metabolic system as a target to reduce the severity of influenza infection. This is facilitated by the fact that, in mammals, copper status can be rapidly reduced by silver nanoparticles and restored after their cancellation.


Subject(s)
Copper/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Ceruloplasmin/physiology , Copper Transport Proteins/metabolism , Copper-Transporting ATPases/physiology , Drug Resistance, Viral , Host-Pathogen Interactions , Humans , Influenza Vaccines , Influenza, Human/drug therapy , Influenza, Human/prevention & control , Influenza, Human/virology , Mammals/metabolism , Metal Nanoparticles/therapeutic use , Molecular Chaperones/metabolism , PrPC Proteins/physiology , RNA, Viral/physiology , Silver/therapeutic use , Superoxide Dismutase-1/physiology , Viral Proteins/physiology , Virus Replication
16.
Gastroenterology ; 156(4): 1173-1189.e5, 2019 03.
Article in English | MEDLINE | ID: mdl-30452922

ABSTRACT

BACKGROUND & AIMS: Wilson disease (WD) is an inherited disorder of copper metabolism that leads to copper accumulation and toxicity in the liver and brain. It is caused by mutations in the adenosine triphosphatase copper transporting ß gene (ATP7B), which encodes a protein that transports copper from hepatocytes into the bile. We studied ATP7B-deficient cells and animals to identify strategies to decrease copper toxicity in patients with WD. METHODS: We used RNA-seq to compare gene expression patterns between wild-type and ATP7B-knockout HepG2 cells exposed to copper. We collected blood and liver tissues from Atp7b-/- and Atp7b+/- (control) rats (LPP) and mice; some mice were given 5 daily injections of an autophagy inhibitor (spautin-1) or vehicle. We obtained liver biopsies from 2 patients with WD in Italy and liver tissues from patients without WD (control). Liver tissues were analyzed by immunohistochemistry, immunofluorescence, cell viability, apoptosis assays, and electron and confocal microscopy. Proteins were knocked down in cell lines using small interfering RNAs. Levels of copper were measured in cell lysates, blood samples, liver homogenates, and subcellular fractions by spectroscopy. RESULTS: After exposure to copper, ATP7B-knockout cells had significant increases in the expression of 103 genes that regulate autophagy (including MAP1LC3A, known as LC3) compared with wild-type cells. Electron and confocal microscopy visualized more autophagic structures in the cytoplasm of ATP7B-knockout cells than wild-type cells after copper exposure. Hepatocytes in liver tissues from patients with WD and from Atp7b-/- mice and rats (but not controls) had multiple autophagosomes. In ATP7B-knockout cells, mammalian target of rapamycin (mTOR) had decreased activity and was dissociated from lysosomes; this resulted in translocation of the mTOR substrate transcription factor EB to the nucleus and activation of autophagy-related genes. In wild-type HepG2 cells (but not ATP7B-knockout cells), exposure to copper and amino acids induced recruitment of mTOR to lysosomes. Pharmacologic inhibitors of autophagy or knockdown of autophagy proteins ATG7 and ATG13 induced and accelerated the death of ATP7B-knockout HepG2 cells compared with wild-type cells. Autophagy protected ATP7B-knockout cells from copper-induced death. CONCLUSION: ATP7B-deficient hepatocytes, such as in those in patients with WD, activate autophagy in response to copper overload to prevent copper-induced apoptosis. Agents designed to activate this autophagic pathway might decrease copper toxicity in patients with WD.


Subject(s)
Apoptosis , Autophagy/genetics , Copper-Transporting ATPases/genetics , Hepatocytes/physiology , Hepatolenticular Degeneration/physiopathology , Liver/physiopathology , Animals , Autophagosomes/ultrastructure , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Benzylamines/pharmacology , Cell Survival , Copper/toxicity , Copper-Transporting ATPases/metabolism , Female , Hep G2 Cells , Hepatocytes/ultrastructure , Humans , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Mitochondria/ultrastructure , Protein Transport , Quinazolines/pharmacology , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
17.
Hum Mol Genet ; 26(6): 1056-1069, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28087733

ABSTRACT

Alterations in mitochondrial activity and morphology have been demonstrated in human cells and tissues from individuals with Down syndrome (DS), as well as in DS mouse models. An impaired activity of the transcriptional coactivator PGC-1α/PPARGC1A due to the overexpression of chromosome 21 genes, such as NRIP1/RIP140, has emerged as an underlying cause of mitochondrial dysfunction in DS. We tested the hypothesis that the activation of the PGC-1α pathway might indeed reverse this mitochondrial dysfunction. To this end, we investigated the effects of metformin, a PGC-1α-activating drug, on mitochondrial morphology and function in DS foetal fibroblasts. Metformin induced both the expression of PGC-1α and an augmentation of its activity, as demonstrated by the increased expression of target genes, strongly promoting mitochondrial biogenesis. Furthermore, metformin enhanced oxygen consumption, ATP production, and overall mitochondrial activity. Most interestingly, this treatment reversed the fragmentation of mitochondria observed in DS and induced the formation of a mitochondrial network with a branched and elongated tubular morphology. Concomitantly, cristae remodelling occurred and the alterations observed by electron microscopy were significantly reduced. We finally demonstrated that the expression of genes of the fission/fusion machinery, namely OPA1 and MFN2, was reduced in trisomic cells and increased by metformin treatment. These results indicate that metformin promotes the formation of a mitochondrial network and corrects the mitochondrial dysfunction in DS cells. We speculate that alterations in the mitochondrial dynamics can be relevant in the pathogenesis of DS and that metformin can efficiently counteract these alterations, thus exerting protective effects against DS-associated pathologies.


Subject(s)
Down Syndrome/genetics , Metformin/administration & dosage , Mitochondria/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Adenosine Triphosphate/biosynthesis , Animals , Disease Models, Animal , Down Syndrome/drug therapy , Down Syndrome/pathology , Fibroblasts/metabolism , GTP Phosphohydrolases/biosynthesis , Humans , Mice , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Oxygen Consumption/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis
18.
Nature ; 501(7465): 116-20, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23913272

ABSTRACT

Newly synthesized proteins and lipids are transported across the Golgi complex via different mechanisms whose respective roles are not completely clear. We previously identified a non-vesicular intra-Golgi transport pathway for glucosylceramide (GlcCer)--the common precursor of the different series of glycosphingolipids-that is operated by the cytosolic GlcCer-transfer protein FAPP2 (also known as PLEKHA8) (ref. 1). However, the molecular determinants of the FAPP2-mediated transfer of GlcCer from the cis-Golgi to the trans-Golgi network, as well as the physiological relevance of maintaining two parallel transport pathways of GlcCer--vesicular and non-vesicular--through the Golgi, remain poorly defined. Here, using mouse and cell models, we clarify the molecular mechanisms underlying the intra-Golgi vectorial transfer of GlcCer by FAPP2 and show that GlcCer is channelled by vesicular and non-vesicular transport to two topologically distinct glycosylation tracks in the Golgi cisternae and the trans-Golgi network, respectively. Our results indicate that the transport modality across the Golgi complex is a key determinant for the glycosylation pattern of a cargo and establish a new paradigm for the branching of the glycosphingolipid synthetic pathway.


Subject(s)
Glucosylceramides/metabolism , Glycosylation , Golgi Apparatus/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biological Transport , Cell Line , Globosides/biosynthesis , Globosides/chemistry , Globosides/metabolism , Glucosylceramides/chemistry , Glycosphingolipids/biosynthesis , Glycosphingolipids/chemistry , Glycosphingolipids/metabolism , Humans , Mice , Mice, Inbred C57BL , Phosphatidylinositol Phosphates/metabolism , trans-Golgi Network/metabolism
19.
Int J Mol Sci ; 21(1)2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31888107

ABSTRACT

Nephropathic cystinosis is a rare lysosomal storage disorder caused by mutations in CTNS gene leading to Fanconi syndrome. Independent studies reported defective clearance of damaged mitochondria and mitochondrial fragmentation in cystinosis. Proteins involved in the mitochondrial dynamics and the mitochondrial ultrastructure were analyzed in CTNS-/- cells treated with cysteamine, the only drug currently used in the therapy for cystinosis but ineffective to treat Fanconi syndrome. CTNS-/- cells showed an overexpression of parkin associated with deregulation of ubiquitination of mitofusin 2 and fission 1 proteins, an altered proteolytic processing of optic atrophy 1 (OPA1), and a decreased OPA1 oligomerization. According to molecular findings, the analysis of electron microscopy images showed a decrease of mitochondrial cristae number and an increase of cristae lumen and cristae junction width. Cysteamine treatment restored the fission 1 ubiquitination, the mitochondrial size, number and lumen of cristae, but had no effect on cristae junction width, making CTNS-/- tubular cells more susceptible to apoptotic stimuli.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Cysteamine/pharmacology , Cystinosis/genetics , Mitochondria/metabolism , Cells, Cultured , Cystinosis/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , GTP Phosphohydrolases/metabolism , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
20.
J Neurosci ; 37(2): 258-268, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28077706

ABSTRACT

Mutations in the genes encoding for gap junction proteins connexin 26 (Cx26) and connexin 30 (Cx30) have been linked to syndromic and nonsyndromic hearing loss in mice and humans. The release of ATP from connexin hemichannels in cochlear nonsensory cells has been proposed to be the main trigger for action potential activity in immature sensory inner hair cells (IHCs), which is crucial for the refinement of the developing auditory circuitry. Using connexin knock-out mice, we show that IHCs fire spontaneous action potentials even in the absence of ATP-dependent intercellular Ca2+ signaling in the nonsensory cells. However, this signaling from nonsensory cells was able to increase the intrinsic IHC firing frequency. We also found that connexin expression is key to IHC functional maturation. In Cx26 conditional knock-out mice (Cx26Sox10-Cre), the maturation of IHCs, which normally occurs at approximately postnatal day 12, was partially prevented. Although Cx30 has been shown not to be required for hearing in young adult mice, IHCs from Cx30 knock-out mice exhibited a comprehensive brake in their development, such that their basolateral membrane currents and synaptic machinery retain a prehearing phenotype. We propose that IHC functional differentiation into mature sensory receptors is initiated in the prehearing cochlea provided that the expression of either connexin reaches a threshold level. As such, connexins regulate one of the most crucial functional refinements in the mammalian cochlea, the disruption of which contributes to the deafness phenotype observed in mice and DFNB1 patients. SIGNIFICANCE STATEMENT: The correct development and function of the mammalian cochlea relies not only on the sensory hair cells, but also on the surrounding nonsensory cells. Although the nonsensory cells have been largely implicated in the general homeostasis in the mature cochlea, their involvement in the initial functional differentiation of the sensory inner hair cells is less clear. Using mutant mouse models for the most common form of congenital deafness in humans, which are knock-outs for the gap-junction channels connexin 26 and connexin 30 genes, we show that defects in nonsensory cells prevented the functional maturation of inner hair cells. In connexin knock-outs, inner hair cells remained stuck at a prehearing stage of development and, as such, are unable to process sound information.


Subject(s)
Cochlea/growth & development , Connexins/physiology , Hair Cells, Auditory, Inner/physiology , Signal Transduction/physiology , Action Potentials/physiology , Animals , Connexin 26 , Connexin 30 , Female , Male , Mice , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL