Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Autophagy ; 19(5): 1551-1561, 2023 05.
Article in English | MEDLINE | ID: mdl-36286616

ABSTRACT

LC3-dependent EV loading and secretion (LDELS) is a secretory autophagy pathway in which the macroautophagy/autophagy machinery facilitates the packaging of cytosolic cargos, such as RNA-binding proteins, into extracellular vesicles (EVs) for secretion outside of the cell. Here, we identify TFRC (transferrin receptor), one of the first proteins found to be secreted via EVs, as a transmembrane cargo of the LDELS pathway. Similar to other LDELS targets, TFRC secretion via EVs genetically requires components of the MAP1LC3/LC3-conjugation machinery but is independent of other ATGs involved in classical autophagosome formation. Furthermore, the packaging and secretion of this transmembrane protein into EVs depends on multiple ESCRT pathway components and the small GTPase RAB27A. Based on these results, we propose that the LDELS pathway promotes TFRC incorporation into EVs and its secretion outside the cell.Abbreviations: ATG: autophagy related; ESCRT: endosomal sorting complexes required for transport; EV: extracellular vesicle; EVP: extracellular vesicle and particle; ILV: intralumenal vesicle; LDELS: LC3-dependent EV loading and secretion; LIR: LC3-interacting region; MVE: multivesicular endosome; RBP: RNA-binding protein; TMT: tandem mass tag; TFRC: transferrin receptor.


Subject(s)
Autophagy , Extracellular Vesicles , Extracellular Vesicles/metabolism , Endosomes/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Receptors, Transferrin/metabolism
2.
J Cell Biol ; 221(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35446347

ABSTRACT

The endolysosome system plays central roles in both autophagic degradation and secretory pathways, including the release of extracellular vesicles and particles (EVPs). Although previous work reveals important interconnections between autophagy and EVP-mediated secretion, our understanding of these secretory events during endolysosome inhibition remains incomplete. Here, we delineate a secretory autophagy pathway upregulated in response to endolysosomal inhibition, which mediates EVP-associated release of autophagic cargo receptors, including p62/SQSTM1. This secretion is highly regulated and dependent on multiple ATGs required for autophagosome formation, as well as the small GTPase Rab27a. Furthermore, disrupting autophagosome maturation, either via genetic inhibition of autophagosome-to-autolysosome fusion or expression of SARS-CoV-2 ORF3a, is sufficient to induce EVP secretion of autophagy cargo receptors. Finally, ATG-dependent EVP secretion buffers against the intracellular accumulation of autophagy cargo receptors when classical autophagic degradation is impaired. Thus, we propose secretory autophagy via EVPs functions as an alternate route to clear sequestered material and maintain proteostasis during endolysosomal dysfunction or impaired autophagosome maturation.


Subject(s)
Autophagy , Extracellular Vesicles , Lysosomes , Proteostasis , Autophagosomes/metabolism , Extracellular Vesicles/metabolism , Humans , Lysosomes/metabolism , SARS-CoV-2 , Sequestosome-1 Protein , Viroporin Proteins , rab27 GTP-Binding Proteins
3.
Cell ; 184(10): 2649-2664.e18, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33848463

ABSTRACT

Receptor tyrosine kinase (RTK)-mediated activation of downstream effector pathways such as the RAS GTPase/MAP kinase (MAPK) signaling cascade is thought to occur exclusively from lipid membrane compartments in mammalian cells. Here, we uncover a membraneless, protein granule-based subcellular structure that can organize RTK/RAS/MAPK signaling in cancer. Chimeric (fusion) oncoproteins involving certain RTKs including ALK and RET undergo de novo higher-order assembly into membraneless cytoplasmic protein granules that actively signal. These pathogenic biomolecular condensates locally concentrate the RAS activating complex GRB2/SOS1 and activate RAS in a lipid membrane-independent manner. RTK protein granule formation is critical for oncogenic RAS/MAPK signaling output in these cells. We identify a set of protein granule components and establish structural rules that define the formation of membraneless protein granules by RTK oncoproteins. Our findings reveal membraneless, higher-order cytoplasmic protein assembly as a distinct subcellular platform for organizing oncogenic RTK and RAS signaling.


Subject(s)
Biomolecular Condensates/metabolism , Cytoplasmic Granules/metabolism , Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Enzyme Activation , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HEK293 Cells , Humans , SOS1 Protein/metabolism , Signal Transduction
5.
iScience ; 20: 14-24, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31546103

ABSTRACT

RNautophagy is a newly described type of selective autophagy whereby cellular RNAs are transported into lysosomes for degradation. This process involves the transmembrane protein SIDT2, which transports double-stranded RNA (dsRNA) across the endolysosomal membrane. We previously demonstrated that SIDT2 is a transcriptional target of p53, but its role in tumorigenesis, if any, is unclear. Unexpectedly, we show here that Sidt2-/- mice with concurrent oncogenic KrasG12D activation develop significantly fewer tumors than littermate controls in a mouse model of lung adenocarcinoma. Consistent with this observation, loss of SIDT2 also leads to enhanced survival and delayed tumor development in an Apcmin/+ mouse model of intestinal cancer. Within the intestine, Apcmin/+;Sidt2-/- mice display accumulation of dsRNA in association with increased phosphorylation of eIF2α and JNK as well as elevated rates of apoptosis. Taken together, our data demonstrate a role for SIDT2, and by extension RNautophagy, in promoting tumor development.

6.
J Immunol ; 202(12): 3483-3492, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31061008

ABSTRACT

dsRNA is a common by-product of viral replication and acts as a potent trigger of antiviral immunity. SIDT1 and SIDT2 are closely related members of the SID-1 transmembrane family. SIDT2 functions as a dsRNA transporter and is required to traffic internalized dsRNA from endocytic compartments into the cytosol for innate immune activation, but the role of SIDT1 in dsRNA transport and in the innate immune response to viral infection is unclear. In this study, we show that Sidt1 expression is upregulated in response to dsRNA and type I IFN exposure and that SIDT1 interacts with SIDT2. Moreover, similar to SIDT2, SIDT1 localizes to the endolysosomal compartment, interacts with the long dsRNA analog poly(I:C), and, when overexpressed, enhances endosomal escape of poly(I:C) in vitro. To elucidate the role of SIDT1 in vivo, we generated SIDT1-deficient mice. Similar to Sidt2-/- mice, SIDT1-deficient mice produced significantly less type I IFN following infection with HSV type 1. In contrast to Sidt2-/- mice, however, SIDT1-deficient animals showed no impairment in survival postinfection with either HSV type 1 or encephalomyocarditis virus. Consistent with this, we observed that, unlike SIDT2, tissue expression of SIDT1 was relatively restricted, suggesting that, whereas SIDT1 can transport extracellular dsRNA into the cytoplasm following endocytosis in vitro, the transport activity of SIDT2 is likely to be functionally dominant in vivo.


Subject(s)
Cardiovirus Infections/immunology , Cytoplasm/metabolism , Encephalomyocarditis virus/physiology , Endosomes/metabolism , Herpes Simplex/immunology , Herpesvirus 1, Human/physiology , Lysosomes/metabolism , Membrane Transport Proteins/metabolism , Nucleotide Transport Proteins/metabolism , Animals , Cells, Cultured , DNA/immunology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotide Transport Proteins/genetics , Poly I-C/immunology , RNA Transport/genetics
7.
Blood ; 133(16): 1729-1741, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30755422

ABSTRACT

Somatically acquired mutations in PHF6 (plant homeodomain finger 6) frequently occur in hematopoietic malignancies and often coincide with ectopic expression of TLX3. However, there is no functional evidence to demonstrate whether these mutations contribute to tumorigenesis. Similarly, the role of PHF6 in hematopoiesis is unknown. We report here that Phf6 deletion in mice resulted in a reduced number of hematopoietic stem cells (HSCs), an increased number of hematopoietic progenitor cells, and an increased proportion of cycling stem and progenitor cells. Loss of PHF6 caused increased and sustained hematopoietic reconstitution in serial transplantation experiments. Interferon-stimulated gene expression was upregulated in the absence of PHF6 in hematopoietic stem and progenitor cells. The numbers of hematopoietic progenitor cells and cycling hematopoietic stem and progenitor cells were restored to normal by combined loss of PHF6 and the interferon α and ß receptor subunit 1. Ectopic expression of TLX3 alone caused partially penetrant leukemia. TLX3 expression and loss of PHF6 combined caused fully penetrant early-onset leukemia. Our data suggest that PHF6 is a hematopoietic tumor suppressor and is important for fine-tuning hematopoietic stem and progenitor cell homeostasis.


Subject(s)
Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Leukemia/etiology , Repressor Proteins/physiology , Animals , Carcinogenesis , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Receptors, Interferon , Repressor Proteins/genetics , Tumor Suppressor Proteins
8.
Bio Protoc ; 8(12): e2890, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-34285999

ABSTRACT

Double-stranded RNA is a potent pathogen-associated molecular pattern (PAMP) produced as a by-product of viral replication and a well-known hallmark of viral infection. Viral dsRNAs can be released from infected cells into the extracellular space and internalized by neighboring cells via endocytosis. Mammals possess multiple pattern recognition receptors (PRRs) capable of detecting viral dsRNAs such as endosomal toll-like receptor 3 (TLR3) and cytosolic RIG-I-like receptors (RLRs) which lead to the production of type I interferons (IFNs). Thus, intracellular localization of viral dsRNA can provide insight into the downstream signaling pathways leading to innate immune activation. Here, we describe a quantitative method for measuring extracellular dsRNA uptake and visualizing subcellular localization of internalized dsRNA via flow cytometry and confocal microscopy respectively.

9.
Methods Mol Biol ; 1714: 237-247, 2018.
Article in English | MEDLINE | ID: mdl-29177867

ABSTRACT

During infection, the cytosolic detection of viral double-stranded RNA (dsRNA) leads to the oligomerization and activation of mitochondrial antiviral signaling protein (MAVS) and the subsequent production of type I interferon (IFN). Here, we describe a novel method of visualizing and quantifying the aggregation of MAVS in response to dsRNA stimulation or viral infection in vitro using confocal microscopy.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Interferon Type I/metabolism , Lentivirus/genetics , Microscopy, Confocal/methods , RNA, Double-Stranded/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/ultrastructure , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , HEK293 Cells , Humans , Mice , Protein Multimerization
10.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28916264

ABSTRACT

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , RNA Transport , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cell Line , Cytoplasm , DEAD Box Protein 58/metabolism , Disease Models, Animal , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Endosomes/metabolism , Female , Gene Expression , Gene Knockout Techniques , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Nucleotide Transport Proteins , Protein Binding , Protein Transport , RNA, Viral/genetics , RNA, Viral/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism
11.
Cell Rep ; 20(3): 668-682, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723569

ABSTRACT

X-linked Inhibitor of Apoptosis (XIAP) deficiency predisposes people to pathogen-associated hyperinflammation. Upon XIAP loss, Toll-like receptor (TLR) ligation triggers RIPK3-caspase-8-mediated IL-1ß activation and death in myeloid cells. How XIAP suppresses these events remains unclear. Here, we show that TLR-MyD88 causes the proteasomal degradation of the related IAP, cIAP1, and its adaptor, TRAF2, by inducing TNF and TNF Receptor 2 (TNFR2) signaling. Genetically, we define that myeloid-specific cIAP1 loss promotes TLR-induced RIPK3-caspase-8 and IL-1ß activity in the absence of XIAP. Importantly, deletion of TNFR2 in XIAP-deficient cells limited TLR-MyD88-induced cIAP1-TRAF2 degradation, cell death, and IL-1ß activation. In contrast to TLR-MyD88, TLR-TRIF-induced interferon (IFN)ß inhibited cIAP1 loss and consequent cell death. These data reveal how, upon XIAP deficiency, a TLR-TNF-TNFR2 axis drives cIAP1-TRAF2 degradation to allow TLR or TNFR1 activation of RIPK3-caspase-8 and IL-1ß. This mechanism may explain why XIAP-deficient patients can exhibit symptoms reminiscent of patients with activating inflammasome mutations.


Subject(s)
Caspase 8/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-1beta/metabolism , Myeloid Differentiation Factor 88/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/metabolism , Toll-Like Receptors/metabolism , Animals , Caspase 8/genetics , Cell Death , Inhibitor of Apoptosis Proteins/deficiency , Inhibitor of Apoptosis Proteins/genetics , Interleukin-1beta/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , TNF Receptor-Associated Factor 2/genetics , Toll-Like Receptors/genetics
12.
Mol Immunol ; 86: 16-22, 2017 06.
Article in English | MEDLINE | ID: mdl-27769523

ABSTRACT

An effective innate immune response relies on the detection of pathogen associated molecular patterns (PAMPs) by various host pattern recognition receptors (PRRs) that result in the production of pro-inflammatory cytokines and chemokines. Viruses and bacteria have co-evolved with the immune system and developed multiple strategies to usurp or circumvent host machinery and blunt the innate immune response in infected cells. Recently, it has become apparent that infected or dying cells can transmit PAMPs and host PRR signalling proteins to uninfected bystander cells to thereby bypass pathogen evasion strategies, and potentiate innate immune signalling. This bystander activation of innate immunity represents an alternative method by which the host can control infections via cell-to-cell communication. In this review, we discuss what is currently known about the intercellular transfer of pathogen- or host-derived RNA, DNA and proteins from infected cells to neighbouring cells and how this impacts on host innate immunity.


Subject(s)
Bystander Effect/immunology , Immunity, Innate , Pathogen-Associated Molecular Pattern Molecules/immunology , Exosomes/metabolism , Humans , Inflammasomes/metabolism , Intercellular Junctions/immunology , Intercellular Junctions/metabolism , Interferon Type I/metabolism , Membrane Proteins/metabolism , Receptors, Pattern Recognition/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Viruses/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...