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










Publication year range
1.
Nat Commun ; 15(1): 974, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38321023

ABSTRACT

Invariant natural killer T (iNKT) cells, a unique T cell population, lend themselves for use as adoptive therapy due to diverse roles in orchestrating immune responses. Originally developed for use in cancer, agenT-797 is a donor-unrestricted allogeneic ex vivo expanded iNKT cell therapy. We conducted an open-label study in virally induced acute respiratory distress syndrome (ARDS) caused by the severe acute respiratory syndrome-2 virus (trial registration NCT04582201). Here we show that agenT-797 rescues exhausted T cells and rapidly activates both innate and adaptive immunity. In 21 ventilated patients including 5 individuals receiving veno-venous extracorporeal membrane oxygenation (VV-ECMO), there are no dose-limiting toxicities. We observe an anti-inflammatory systemic cytokine response and infused iNKT cells are persistent during follow-up, inducing only transient donor-specific antibodies. Clinical signals of associated survival and prevention of secondary infections are evident. Cellular therapy using off-the-shelf iNKT cells is safe, can be rapidly scaled and is associated with an anti-inflammatory response. The safety and therapeutic potential of iNKT cells across diseases including infections and cancer, warrants randomized-controlled trials.


Subject(s)
Natural Killer T-Cells , Neoplasms , Respiratory Distress Syndrome , Humans , Cytokines/metabolism , Anti-Inflammatory Agents
2.
J Immunol ; 211(4): 633-647, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37449888

ABSTRACT

NK cells and CD8 T cells use cytotoxic molecules to kill virally infected and tumor cell targets. While perforin and granzyme B (GzmB) are the most commonly studied lytic molecules, less is known about granzyme K (GzmK). However, this granzyme has been recently associated with improved prognosis in solid tumors. In this study, we show that, in humans, GzmK is predominantly expressed by innate-like lymphocytes, as well as a newly identified population of GzmK+CD8+ non- mucosal-associated invariant T cells with innate-like characteristics. We found that GzmK+ T cells are KLRG1+EOMES+IL-7R+CD62L-Tcf7int, suggesting that they are central memory T and effector memory T cells. Furthermore, GzmK+ cells are absent/low in cord blood, suggesting that GzmK is upregulated with immune experience. Surprisingly, GzmK+ cells respond to cytokine stimuli alone, whereas TCR stimulation downregulates GzmK expression, coinciding with GzmB upregulation. GzmK+ cells have reduced IFN-γ production compared with GzmB+ cells in each T cell lineage. Collectively, this suggests that GzmK+ cells are not naive, and they may be an intermediate memory-like or preterminally differentiated population. GzmK+ cells are enriched in nonlymphoid tissues such as the liver and adipose. In colorectal cancer, GzmK+ cells are enriched in the tumor and can produce IFN-γ, but GzmK+ expression is mutually exclusive with IL-17a production. Thus, in humans, GzmK+ cells are innate memory-like cells that respond to cytokine stimulation alone and may be important effector cells in the tumor.


Subject(s)
CD8-Positive T-Lymphocytes , Cytokines , Granzymes , Humans , Cytokines/metabolism , Granzymes/metabolism , Killer Cells, Natural , Receptors, Antigen, T-Cell/metabolism
4.
Nat Commun ; 14(1): 3763, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353482

ABSTRACT

Altered protein phosphorylation in cancer cells often leads to surface presentation of phosphopeptide neoantigens. However, their role in cancer immunogenicity remains unclear. Here we describe a mechanism by which an HLA-B*0702-specific acute myeloid leukemia phosphoneoantigen, pMLL747-755 (EPR(pS)PSHSM), is recognized by a cognate T cell receptor named TCR27, a candidate for cancer immunotherapy. We show that the replacement of phosphoserine P4 with serine or phosphomimetics does not affect pMHC conformation or peptide-MHC affinity but abrogates TCR27-dependent T cell activation and weakens binding between TCR27 and pMHC. Here we describe the crystal structures for TCR27 and cognate pMHC, map of the interface produced by nuclear magnetic resonance, and a ternary complex generated using information-driven protein docking. Our data show that non-covalent interactions between the epitope phosphate group and TCR27 are crucial for TCR specificity. This study supports development of new treatment options for cancer patients through target expansion and TCR optimization.


Subject(s)
Phosphopeptides , Receptors, Antigen, T-Cell , Humans , Phosphopeptides/metabolism , Protein Binding
5.
Nat Commun ; 10(1): 4068, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31492851

ABSTRACT

The aldehyde dehydrogenase (ALDH) family of metabolic enzymes converts aldehydes to carboxylates. Here, we find that the reductive consequence of ALDH7A1 activity, which generates NADH (nicotinamide adenine dinucleotide, reduced form) from NAD, underlies how ALDH7A1 coordinates a broad inhibition of the intracellular transport pathways. Studying vesicle formation by the Coat Protein I (COPI) complex, we elucidate that NADH generated by ALDH7A1 targets Brefeldin-A ADP-Ribosylated Substrate (BARS) to inhibit COPI vesicle fission. Moreover, defining a physiologic role for the broad transport inhibition exerted by ALDH7A1, we find that it acts to reduce energy consumption during hypoxia and starvation to promote cellular energy homeostasis. These findings advance the understanding of intracellular transport by revealing how the coordination of multiple pathways can be achieved, and also defining circumstances when such coordination is needed, as well as uncovering an unexpected way that NADH acts in cellular energetics.


Subject(s)
Alcohol Oxidoreductases/metabolism , Aldehyde Dehydrogenase/metabolism , DNA-Binding Proteins/metabolism , Energy Metabolism , Homeostasis , Intracellular Space/metabolism , Alcohol Oxidoreductases/genetics , Aldehyde Dehydrogenase/genetics , Biological Transport , COP-Coated Vesicles/metabolism , Cell Hypoxia , DNA-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , NAD/metabolism , Signal Transduction , Starvation
6.
Nat Immunol ; 19(12): 1330-1340, 2018 12.
Article in English | MEDLINE | ID: mdl-30420624

ABSTRACT

Up to 49% of certain types of cancer are attributed to obesity, and potential mechanisms include overproduction of hormones, adipokines, and insulin. Cytotoxic immune cells, including natural killer (NK) cells and CD8+ T cells, are important in tumor surveillance, but little is known about the impact of obesity on immunosurveillance. Here, we show that obesity induces robust peroxisome proliferator-activated receptor (PPAR)-driven lipid accumulation in NK cells, causing complete 'paralysis' of their cellular metabolism and trafficking. Fatty acid administration, and PPARα and PPARδ (PPARα/δ) agonists, mimicked obesity and inhibited mechanistic target of rapamycin (mTOR)-mediated glycolysis. This prevented trafficking of the cytotoxic machinery to the NK cell-tumor synapse. Inhibiting PPARα/δ or blocking the transport of lipids into mitochondria reversed NK cell metabolic paralysis and restored cytotoxicity. In vivo, NK cells had blunted antitumor responses and failed to reduce tumor growth in obesity. Our results demonstrate that the lipotoxic obese environment impairs immunosurveillance and suggest that metabolic reprogramming of NK cells may improve cancer outcomes in obesity.


Subject(s)
Immunologic Surveillance/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Melanoma, Experimental/immunology , Obesity/immunology , Adult , Animals , Female , Humans , Killer Cells, Natural/pathology , Male , Melanoma, Experimental/complications , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/complications , Young Adult
7.
J Immunol ; 200(9): 3160-3169, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29592961

ABSTRACT

Mycobacterium tuberculosis is an extremely successful pathogen, and its success is widely attributed to its ability to manipulate the intracellular environment of macrophages. A central phenomenon of tuberculosis pathology enabling immune evasion is the capacity of virulent M. tuberculosis (H37Rv) to induce macrophage necrosis, which facilitates the escape of the mycobacteria from the macrophage and spread of infection. In contrast, avirulent M. tuberculosis (H37Ra) induces macrophage apoptosis, which permits Ag presentation and activation of adaptive immunity. Previously, we found that H37Rv induces plasma membrane microdisruptions, leading to necrosis in the absence of plasma membrane repair. In contrast, H37Ra permits plasma membrane repair, which changes the host cell death modality to apoptosis, suggesting that membrane repair is critical for sequestering the pathogen in apoptotic vesicles. However, mechanisms of plasma membrane repair induced in response to M. tuberculosis infection remain unknown. Plasma membrane repair is known to induce a Ca2+-mediated signaling, which recruits lysosomes to the area of damaged plasma membrane sites for its resealing. In this study, we found that the small GTPase Arl8b is required for plasma membrane repair by controlling the exocytosis of lysosomes in cell lines and in human primary macrophages. Importantly, we found that the Arl8b secretion pathway is crucial to control the type of cell death of the M. tuberculosis-infected macrophages. Indeed, Arl8b-depleted macrophages infected with avirulent H37Ra undergo necrotic instead of apoptotic cell death. These findings suggest that membrane repair mediated by Arl8b may be an important mechanism distinguishing avirulent from virulent M. tuberculosis-induced necrotic cell death.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Membrane/metabolism , Lysosomes/metabolism , Macrophages/microbiology , Tuberculosis/metabolism , Apoptosis/physiology , Humans , Immune Evasion/physiology , Macrophages/metabolism , Mycobacterium tuberculosis/pathogenicity , Necrosis/metabolism , Necrosis/microbiology , Virulence/physiology
8.
J Clin Invest ; 127(9): 3300-3312, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28758901

ABSTRACT

M2 macrophages, innate lymphoid type 2 cells (ILC2s), eosinophils, Tregs, and invariant NK T cells (iNKT cells) all help to control adipose tissue inflammation, while M1 macrophages, TNF, and other inflammatory cytokines drive inflammation and insulin resistance in obesity. Stromal cells regulate leukocyte responses in lymph nodes, but the role of stromal cells in adipose tissue inflammation is unknown. PDGFRα+ stromal cells are major producers of IL-33 in adipose tissue. Here, we show that mesenchymal cadherin-11 modulates stromal fibroblast function. Cadherin-11-deficient mice displayed increased stromal production of IL-33, with concomitant enhancements in ILC2s and M2 macrophages that helped control adipose tissue inflammation. Higher expression levels of IL-33 in cadherin-11-deficient mice mediated ILC2 activation, resulting in higher IL-13 expression levels and M2 macrophage expansion in adipose tissue. Consistent with reduced adipose tissue inflammation, cadherin-11-deficient mice were protected from obesity-induced glucose intolerance and adipose tissue fibrosis. Importantly, anti-cadherin-11 mAb blockade similarly improved inflammation and glycemic control in obese WT mice. These results suggest that stromal fibroblasts expressing cadherin-11 regulate adipose tissue inflammation and thus highlight cadherin-11 as a potential therapeutic target for the management of obesity.


Subject(s)
Adipose Tissue/physiopathology , Cadherins/metabolism , Diabetes Mellitus, Experimental/physiopathology , Inflammation/physiopathology , Adipocytes/cytology , Adipose Tissue/metabolism , Animals , Cell Differentiation , Crosses, Genetic , Diabetes Mellitus, Experimental/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Glucose Intolerance/metabolism , Inflammation/metabolism , Insulin Resistance , Interleukin-13/metabolism , Interleukin-33/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Obesity/metabolism , Phenotype
9.
Immunity ; 46(2): 273-286, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228283

ABSTRACT

Adipose tissue has a dynamic immune system that adapts to changes in diet and maintains homeostatic tissue remodeling. Adipose type 1 innate lymphoid cells (AT1-ILCs) promote pro-inflammatory macrophages in obesity, but little is known about their functions at steady state. Here we found that human and murine adipose tissue harbor heterogeneous populations of AT1-ILCs. Experiments using parabiotic mice fed a high-fat diet (HFD) showed differential trafficking of AT1-ILCs, particularly in response to short- and long-term HFD and diet restriction. At steady state, AT1-ILCs displayed cytotoxic activity toward adipose tissue macrophages (ATMs). Depletion of AT1-ILCs and perforin deficiency resulted in alterations in the ratio of inflammatory to anti-inflammatory ATMs, and adoptive transfer of AT1-ILCs exacerbated metabolic disorder. Diet-induced obesity impaired AT1-ILC killing ability. Our findings reveal a role for AT1-ILCs in regulating ATM homeostasis through cytotoxicity and suggest that this function is relevant in both homeostasis and metabolic disease.


Subject(s)
Adipose Tissue/immunology , Cytotoxicity, Immunologic/immunology , Homeostasis/immunology , Lymphocytes/immunology , Macrophages/immunology , Obesity/immunology , Adipose Tissue/cytology , Animals , Female , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Obesity/pathology
10.
J Cell Biol ; 213(6): 631-40, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27325790

ABSTRACT

Lysosome exocytosis plays a major role in resealing plasma membrane (PM) disruptions. This process involves two sequential steps. First, lysosomes are recruited to the periphery of the cell and then fuse with the damaged PM. However, the trafficking molecular machinery involved in lysosome exocytosis and PM repair (PMR) is poorly understood. We performed a systematic screen of the human Rab family to identify Rabs required for lysosome exocytosis and PMR. Rab3a, which partially localizes to peripheral lysosomes, was one of the most robust hits. Silencing of Rab3a or its effector, synaptotagmin-like protein 4a (Slp4-a), leads to the collapse of lysosomes to the perinuclear region and inhibition of PMR. Importantly, we have also identified a new Rab3 effector, nonmuscle myosin heavy chain IIA, as part of the complex formed by Rab3a and Slp4-a that is responsible for lysosome positioning at the cell periphery and lysosome exocytosis.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/physiology , Lysosomes/metabolism , Lysosomes/physiology , rab3A GTP-Binding Protein/metabolism , Cell Line , Cell Line, Tumor , Exocytosis/physiology , HEK293 Cells , HeLa Cells , Humans , Leukocytes, Mononuclear , Myosin Heavy Chains/metabolism , Vesicular Transport Proteins/metabolism
11.
J Cell Sci ; 129(7): 1490-9, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26906413

ABSTRACT

The sarcoplasmic reticulum is a network of tubules and cisternae localized in close association with the contractile apparatus, and regulates Ca(2+)dynamics within striated muscle cell. The sarcoplasmic reticulum maintains its shape and organization despite repeated muscle cell contractions, through mechanisms which are still under investigation. The ESCRT complexes are essential to organize membrane subdomains and modify membrane topology in multiple cellular processes. Here, we report for the first time that ESCRT-II proteins play a role in the maintenance of sarcoplasmic reticulum integrity inC. elegans ESCRT-II proteins colocalize with the sarcoplasmic reticulum marker ryanodine receptor UNC-68. The localization at the sarcoplasmic reticulum of ESCRT-II and UNC-68 are mutually dependent. Furthermore, the characterization of ESCRT-II mutants revealed a fragmentation of the sarcoplasmic reticulum network, associated with an alteration of Ca(2+)dynamics. Our data provide evidence that ESCRT-II proteins are involved in sarcoplasmic reticulum shaping.


Subject(s)
Caenorhabditis elegans/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Muscle Cells/metabolism , Muscle Contraction/physiology , Sarcoplasmic Reticulum/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
12.
J Immunol ; 194(5): 2079-88, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25637027

ABSTRACT

Dendritic cells (DCs) are specialized APCs with the ability to prime naive T cells. DCs first sample Ags from the environment and then orchestrate their processing and loading onto MHC class II (MHC II) Ag-presenting molecules in lysosomes. Once MHC II molecules have bound a peptide, the MHC II-peptide complex is delivered to the cell surface for presentation to CD4(+) T cells. Regulation of Ag uptake via macropinocytosis and phagocytosis has been extensively studied, as well as trafficking in early endocytic vesicles notably regulated by the small GTPase Rab5 and its effectors. However, little is known about the regulators of Ag delivery from early endosomes to lysosomal compartments where the proper pH, proteases, MHC II, invariant chain, and HLA-DM reside, awaiting exogenous Ags for loading. In this article, we report the crucial role of the small GTPase ADP-ribosylation factor-like 8b (Arl8b) in MHC II presentation in DCs. We show for the first time, to our knowledge, that Arl8b localizes to MHC II compartments in DCs and regulates formation of MHC II-peptide complexes. Arl8b-silenced DCs display a defect in MHC II-Ag complex formation and its delivery to the cell surface during infection resulting in a defect in T cell recognition. Our results highlight the role of Arl8b as a trafficking regulator of the late stage of complex formation and MHC II presentation in DCs.


Subject(s)
ADP-Ribosylation Factors/immunology , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Lysosomes/immunology , ADP-Ribosylation Factors/antagonists & inhibitors , ADP-Ribosylation Factors/genetics , Animals , Antigens/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Line , Chickens , Dendritic Cells/cytology , Endosomes/immunology , Gene Expression Regulation , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Primary Cell Culture , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Spleen/cytology , Spleen/immunology
13.
Nat Immunol ; 16(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25436972

ABSTRACT

Invariant natural killer T cells (iNKT cells) are lipid-sensing innate T cells that are restricted by the antigen-presenting molecule CD1d and express the transcription factor PLZF. iNKT cells accumulate in adipose tissue, where they are anti-inflammatory, but the factors that contribute to their anti-inflammatory nature, as well as their targets in adipose tissue, are unknown. Here we found that iNKT cells in adipose tissue had a unique transcriptional program and produced interleukin 2 (IL-2) and IL-10. Unlike other iNKT cells, they lacked PLZF but expressed the transcription factor E4BP4, which controlled their IL-10 production. The adipose iNKT cells were a tissue-resident population that induced an anti-inflammatory phenotype in macrophages and, through the production of IL-2, controlled the number, proliferation and suppressor function of regulatory T cells (Treg cells) in adipose tissue. Thus, iNKT cells in adipose tissue are unique regulators of immunological homeostasis in this tissue.


Subject(s)
Adipose Tissue/immunology , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/immunology , Natural Killer T-Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Adipose Tissue/cytology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cell Growth Processes/immunology , Female , Flow Cytometry , Gene Expression Regulation , Homeostasis/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Macrophages/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
14.
Mol Biol Cell ; 24(23): 3721-35, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24088571

ABSTRACT

Natural killer (NK) lymphocytes contain lysosome-related organelles (LROs), known as lytic granules, which upon formation of immune synapse with the target cell, polarize toward the immune synapse to deliver their contents to the target cell membrane. Here, we identify a small GTP-binding protein, ADP-ribosylation factor-like 8b (Arl8b), as a critical factor required for NK cell-mediated cytotoxicity. Our findings indicate that Arl8b drives the polarization of lytic granules and microtubule-organizing centers (MTOCs) toward the immune synapse between effector NK lymphocytes and target cells. Using a glutathione S-transferase pull-down approach, we identify kinesin family member 5B (KIF5B; the heavy chain of kinesin-1) as an interaction partner of Arl8b from NK cell lysates. Previous studies showed that interaction between kinesin-1 and Arl8b is mediated by SifA and kinesin-interacting protein (SKIP) and the tripartite complex drives the anterograde movement of lysosomes. Silencing of both KIF5B and SKIP in NK cells, similar to Arl8b, led to failure of MTOC-lytic granule polarization to the immune synapse, suggesting that Arl8b and kinesin-1 together control this critical step in NK cell cytotoxicity.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic , GTP Phosphohydrolases/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Adaptor Proteins, Signal Transducing/metabolism , Cytoplasmic Granules/ultrastructure , Exocytosis , Gene Silencing , HeLa Cells , Humans , Immunological Synapses/metabolism , Immunological Synapses/ultrastructure , Killer Cells, Natural/ultrastructure , Kinesins/metabolism , Microtubule-Organizing Center/metabolism , Protein Transport
15.
J Cell Sci ; 125(Pt 3): 685-94, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22389403

ABSTRACT

Endosomes and autophagosomes are two vesicular compartments involved in the degradation and recycling of cellular material. They both undergo a maturation process and finally fuse with the lysosome. In mammals, the convergence between endosomes and autophagosomes is a multistep process that can generate intermediate vesicles named amphisomes. Using knockdowns and mutants of the ESCRT machinery (ESCRT-0-ESCRT-III, ATPase VPS-4) and the autophagic pathway (LGG-1, LGG-2, ATG-7, TOR), we analyzed in vivo the functional links between endosomal maturation and autophagy in Caenorhabditis elegans. We report here that, despite a strong heterogeneity of their developmental phenotypes, all ESCRT mutants present an accumulation of abnormal endosomes and autophagosomes. We show that this accumulation of autophagosomes is secondary to the formation of enlarged endosomes and is due to the induction of the autophagic flux and not a blockage of fusion with lysosomes. We demonstrate that the induction of autophagy is not responsible for the lethality of ESCRT mutants but has a protective role on cellular degradation. We also show that increasing the basal level of autophagy reduces the formation of enlarged endosomes in ESCRT mutants. Together, our data indicate that the induction of autophagy is a protective response against the formation of an abnormal vesicular compartment.


Subject(s)
Autophagy/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Adaptation, Physiological , Animals , Animals, Genetically Modified , Autophagy/physiology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Survival/genetics , Cell Survival/physiology , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Gene Knockdown Techniques , Genes, Helminth , Mutation , Phenotype , RNA Interference
16.
Worm ; 1(4): 216-20, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-24058852

ABSTRACT

The endosomal and autophagic pathways are essential for the degradation and renewal of cellular components. After a complex maturation process, both pathways converge to their final destination, the lysosome. A close link between these two pathways was described along the last decade, notably through the analysis of ESCRT mutants. Although in mammals ESCRT mutants are unable to complete autophagic maturation due to the lack of fusion with the endolysosomal system, the role of ESCRT in the autophagic process still remains an open issue. Using C. elegans, we recently showed that blockage of the endosomal maturation triggers the induction of autophagic activity in ESCRT mutant.(1) This increase of autophagic flux is an attempt to correct cellular defects and promote the survival of mutant animals.

17.
Autophagy ; 6(5): 622-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20523114

ABSTRACT

Autophagy has an important function in degrading cytoplasmic components to maintain cellular homeostasis, but is also required during development. The formation of the autophagic vesicles requires the recruitment of the Atg8 ubiquitin-like proteins to the membrane of the nascent autophagosomes. Atg8 is a highly conserved gene which has been duplicated during metazoan evolution. In this report we have investigated, in the nematode C. elegans, the functions and localizations of the two Atg8p homologues LGG-2 and LGG-1. Phylogenetic analyses suggest that LGG-2 is more closely related to the human protein LC3 than LGG-1. LGG-1 but not LGG-2 is able to functionally complement the atg8 mutant yeast. The C-terminal glycine residue of LGG-2 is essential for post-translational modification and localization to the autophagosomes. During C. elegans development the two proteins share a similar expression pattern and localization but LGG-2 is more abundant in the neurons. Using genetic tools to either reduce or increase the autophagic flux we show that both LGG-2 and LGG-1 are addressed to the autophagosomal/lysosomal degradative system. We also demonstrate that the localization of both proteins is modified in several physiological processes when autophagy is induced, namely during diapause "dauer" larval formation, starvation and aging. Finally, we demonstrate that both LGG-2 and LGG-1 act synergistically and are involved in dauer formation and longevity of the worm.


Subject(s)
Autophagy , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/physiology , Longevity/physiology , Phagosomes/metabolism , Amino Acid Sequence , Animals , Autophagy/genetics , Autophagy-Related Protein 8 Family , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Helminth/genetics , Genome/genetics , Glycine/metabolism , Humans , Larva/physiology , Longevity/genetics , Lysosomes/metabolism , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Mutation/genetics , Protein Transport , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Nucleic Acid
18.
Biol Cell ; 102(3): 191-202, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20059450

ABSTRACT

ESCRTs (endosomal sorting complexes required for transport) were first discovered in yeast and are known to be required in the biogenesis of the MVB (multivesicular body). Most ESCRT research has been carried out in vitro using models such as yeast and mammalian cells in culture. The role of the ESCRTs genes in endosome maturation is conserved from yeast to mammals, but little is known about their function during development in multicellular organisms. Since ESCRTs play a leading role in regulating some cell signalling pathways by addressing receptors to the lysosome, it appears important to monitor ESCRT functions in multicellular models. The present review summarizes recent research on the developmental and cellular functions of the ESCRT in Caenorhabditis elegans, Drosophila melanogaster, Mus musculus or Arabidopsis thaliana.


Subject(s)
Cell Differentiation/physiology , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Signal Transduction/physiology , Animals , Endosomal Sorting Complexes Required for Transport/ultrastructure , Endosomes/ultrastructure , Evolution, Molecular , Gene Expression Regulation, Developmental/physiology , Lysosomes/ultrastructure , Models, Animal , Multivesicular Bodies/metabolism , Multivesicular Bodies/ultrastructure , Protein Transport/physiology
19.
FEBS J ; 276(18): 5030-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19663909

ABSTRACT

To elucidate the function of kappa class glutathione transferases (GSTs) in multicellular organisms, their expression and silencing were investigated in Caenorhabditis elegans. In contrast with most vertebrates, which possess only one GST kappa gene, two distinct genes encoding GSTK-1 and GSTK-2 are present in the C. elegans genome. The amino acid sequences of GSTK-1 and GSTK-2 share around 30% similarity with the human hGSTK1 sequence and, like the human transferase, GSTK-1 contains a C-terminal peroxisomal targeting sequence. gstk-1 and gstk-2 genes show distinct developmental and tissue expression patterns. We show that GSTK-2 is localized in the mitochondria and expressed mainly in the pharynx, muscles and epidermis, whereas GSTK-1 is restricted to peroxisomes and expressed in the intestine, body wall muscles and epidermis. In order to determine the potential role(s) of GST kappa genes in C. elegans, specific silencing of the gstk-1 and gstk-2 genes was performed by an RNA interference approach. Knockdown of gstk-1 or gstk-2 had no apparent effect on C. elegans reproduction, development, locomotion or lifespan. By contrast, when biological functions (oxygen consumption and lipid metabolism) related to peroxisomes and/or mitochondria were investigated, we observed a significant decrease in respiration rate and a lower concentration of the monounsaturated fatty acid cis-vaccenic acid (18:1omega7) when worms were fed on bacteria expressing RNA interference targeting both gstk-1 and gstk-2. These results demonstrate that GST kappa, although not essential for the worm's life, may be involved in energetic and lipid metabolism, two functions related to mitochondria and peroxisomes.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Glutathione Transferase/physiology , Lipid Metabolism , Mitochondria/enzymology , Oxygen Consumption , Peroxisomes/enzymology , Animals , Glutathione Transferase/analysis , Glutathione Transferase/genetics
20.
Biol Cell ; 101(10): 599-615, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19432559

ABSTRACT

BACKGROUND INFORMATION: Within the endocytic pathway, the ESCRT (endosomal sorting complex required for transport) machinery is essential for the biogenesis of MVBs (multivesicular bodies). In yeast, ESCRTs are recruited at the endosomal membrane and are involved in cargo sorting into intralumenal vesicles of the MVBs. RESULTS: In the present study, we characterize the ESCRT-III protein CeVPS-32 (Caenorhabditis elegans vacuolar protein sorting 32) and its interactions with CeVPS-27, CeVPS-23 and CeVPS-4. In contrast with other CevpsE (class E vps) genes, depletion of Cevps-32 is embryonic lethal with severe defects in the remodelling of epithelial cell shape during organogenesis. Furthermore, Cevps-32 animals display an accumulation of enlarged early endosomes in epithelial cells and an accumulation of autophagosomes. The CeVPS-32 protein is enriched in epithelial tissues and in residual bodies during spermatid maturation. We show that CeVPS-32 and CeVPS-27/Hrs (hepatocyte-growth-factor-regulated tyrosine kinase substrate) are enriched in distinct subdomains at the endosomal membrane. CeVPS-27-positive subdomains are also enriched for the ESCRT-I protein CeVPS-23/TSG101 (tumour susceptibility gene 101). The formation of CeVPS-27 subdomains is not affected by the depletion of CeVPS-23, CeVPS-32 or the ATPase CeVPS-4. CONCLUSION: Our results suggest that the formation of membrane subdomains is essential for the maturation of endosomes.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Endosomes/metabolism , Epithelial Cells/metabolism , Vesicular Transport Proteins/metabolism , Animals , Autophagy/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/chemistry , Disorders of Sex Development , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Embryonic Development , Genes, Lethal , Genes, Reporter , Germ Cells/metabolism , Immunohistochemistry , Larva/growth & development , Larva/metabolism , Larva/ultrastructure , Life Cycle Stages , Male , Membranes/metabolism , Organogenesis , RNA Interference , Sex Characteristics , Transfection , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...