Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38903088

ABSTRACT

The bone-resorbing activity of osteoclasts plays a critical role in the life-long remodeling of our bones that is perturbed in many bone loss diseases. Multinucleated osteoclasts are formed by the fusion of precursor cells, and larger cells - generated by an increased number of cell fusion events - have higher resorptive activity. We find that osteoclast fusion and bone-resorption are promoted by reactive oxygen species (ROS) signaling and by an unconventional low molecular weight species of La protein, located at the osteoclast surface. Here, we develop the hypothesis that La's unique regulatory role in osteoclast multinucleation and function is controlled by a ROS switch in La trafficking. Using antibodies that recognize reduced or oxidized species of La, we find that differentiating osteoclasts enrich an oxidized species of La at the cell surface, which is distinct from the reduced La species conventionally localized within cell nuclei. ROS signaling triggers the shift from reduced to oxidized La species, its dephosphorylation and delivery to the surface of osteoclasts, where La promotes multinucleation and resorptive activity. Moreover, intracellular ROS signaling in differentiating osteoclasts oxidizes critical cysteine residues in the C-terminal half of La, producing this unconventional La species that promotes osteoclast fusion. Our findings suggest that redox signaling induces changes in the location and function of La and may represent a promising target for novel skeletal therapies.

2.
Bone Res ; 12(1): 10, 2024 02 20.
Article in English | MEDLINE | ID: mdl-38378678

ABSTRACT

Fibrous dysplasia (FD) is a rare, disabling skeletal disease for which there are no established treatments. Growing evidence supports inhibiting the osteoclastogenic factor receptor activator of nuclear kappa-B ligand (RANKL) as a potential treatment strategy. In this study, we investigated the mechanisms underlying RANKL inhibition in FD tissue and its likely indirect effects on osteoprogenitors by evaluating human FD tissue pre- and post-treatment in a phase 2 clinical trial of denosumab (NCT03571191) and in murine in vivo and ex vivo preclinical models. Histological analysis of human and mouse tissue demonstrated increased osteogenic maturation, reduced cellularity, and reduced expression of the pathogenic Gαs variant in FD lesions after RANKL inhibition. RNA sequencing of human and mouse tissue supported these findings. The interaction between osteoclasts and mutant osteoprogenitors was further assessed in an ex vivo lesion model, which indicated that the proliferation of abnormal FD osteoprogenitors was dependent on osteoclasts. The results from this study demonstrated that, in addition to its expected antiosteoclastic effect, denosumab reduces FD lesion activity by decreasing FD cell proliferation and increasing osteogenic maturation, leading to increased bone formation within lesions. These findings highlight the unappreciated role of cellular crosstalk between osteoclasts and preosteoblasts/osteoblasts as a driver of FD pathology and demonstrate a novel mechanism of action of denosumab in the treatment of bone disease.TRIAL REGISTRATION: ClinicalTrials.gov NCT03571191.


Subject(s)
Denosumab , Fibrous Dysplasia of Bone , Animals , Humans , Mice , Denosumab/pharmacology , Fibrous Dysplasia of Bone/drug therapy , Ligands , Osteoblasts/metabolism , Osteogenesis/genetics
3.
iScience ; 26(4): 106470, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37091244

ABSTRACT

Elucidating a basic blueprint of osteoclast-osteoblast coordination in skeletal remodeling and understanding how this coordination breaks down with age and disease is essential for addressing the growing skeletal health problem in our aging population. The paucity of simple, activatable, biologically relevant models of osteoclast-osteoblast coordination has hindered our understanding of how skeletal remolding is regulated. Here, we describe an inducible ex vivo model of osteoclast-osteoblast progenitor coordination. Induction activates the release of osteoclastogenic factors from osteoprogenitors, which elicits the differentiation and fusion of neighboring preosteoclasts. In turn, multinucleated osteoclasts release soluble coupling factors, RANK+ extracellular vesicles and promote osteoprogenitor proliferation, recapitulating aspects of perturbed coordination in diseases underpinned by excessive osteoclast formation. We expect this model to expedite the investigation of cell-cell fusion, osteoclast-osteoblast progenitor coordination, and extracellular vesicle signaling during bone remodeling and offer a powerful tool for evaluating signaling cascades and novel therapeutic interventions in osteoclast-linked skeletal disease.

4.
Nat Commun ; 14(1): 616, 2023 02 04.
Article in English | MEDLINE | ID: mdl-36739273

ABSTRACT

Multinucleated osteoclasts, essential for skeletal remodeling in health and disease, are formed by the fusion of osteoclast precursors, where each fusion event raises their bone-resorbing activity. Here we show that the nuclear RNA chaperone, La protein has an additional function as an osteoclast fusion regulator. Monocyte-to-osteoclast differentiation starts with a drastic decrease in La levels. As fusion begins, La reappears as a low molecular weight species at the osteoclast surface, where it promotes fusion. La's role in promoting osteoclast fusion is independent of canonical La-RNA interactions and involves direct interactions between La and Annexin A5, which anchors La to transiently exposed phosphatidylserine at the surface of fusing osteoclasts. Disappearance of cell-surface La, and the return of full length La to the nuclei of mature, multinucleated osteoclasts, acts as an off switch of their fusion activity. Targeting surface La in a novel explant model of fibrous dysplasia inhibits excessive osteoclast formation characteristic of this disease, highlighting La's potential as a therapeutic target.


Subject(s)
Bone Resorption , Osteogenesis , Humans , Bone Resorption/metabolism , Cell Differentiation , Cell Fusion , Cell Membrane/metabolism , Membrane Proteins/metabolism , Osteoclasts/metabolism
5.
Proc Natl Acad Sci U S A ; 119(38): e2202490119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36095199

ABSTRACT

Muscle cell fusion is a multistep process where the final step of the reaction drives progression beyond early hemifusion events to complete fusion. This step requires activity of the muscle-specific fusogen Myomerger, a single-pass transmembrane protein containing 84 amino acids with an ectodomain that includes two α-helices. Previous studies have demonstrated that Myomerger acts by destabilizing membranes through generation of elastic stresses in the outer leaflet of the plasma membrane. An obvious question is how such destabilizing activity might be regulated to avoid membrane and cellular damage, and how the two juxtaposed helices cooperate in fusion. Using cellular fusion assays and in vitro liposome assays, we report that the two helices possess unique characteristics, both of which are needed for full activity of the protein. We demonstrate that externalized phosphatidylserine (PS), a lipid previously implicated in myoblast fusion, has a determinant role in the regulation of Myomerger activity. The membrane-proximal, amphipathic Helix-1 is normally disordered and its α-helical structure is induced by PS, making membrane interactions more efficacious. The distal, more hydrophobic Helix-2 is intrinsically ordered, possesses an ability to insert into membranes, and augments the membrane-stressing effects of Helix-1. These data reveal that Myomerger fusogenic activity is an exquisitely orchestrated event involving its two ectodomain helices, which are controlled by membrane lipid composition, providing an explanation as to how its membrane-stressing activity is spatially and temporally regulated during the final step of myoblast fusion.


Subject(s)
Cell Fusion , Membrane Proteins , Myoblasts , Phosphatidylserines , Animals , Cell Line , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myoblasts/physiology
6.
J Biol Chem ; 296: 100411, 2021.
Article in English | MEDLINE | ID: mdl-33581114

ABSTRACT

Formations of myofibers, osteoclasts, syncytiotrophoblasts, and fertilized zygotes share a common step, cell-cell fusion. Recent years have brought about considerable progress in identifying some of the proteins involved in these and other cell-fusion processes. However, even for the best-characterized cell fusions, we still do not know the mechanisms that regulate the timing of cell-fusion events. Are they fully controlled by the expression of fusogenic proteins or do they also depend on some triggering signal that activates these proteins? The latter scenario would be analogous to the mechanisms that control the timing of exocytosis initiated by Ca2+ influx and virus-cell fusion initiated by low pH- or receptor interaction. Diverse cell fusions are accompanied by the nonapoptotic exposure of phosphatidylserine at the surface of fusing cells. Here we review data on the dependence of membrane remodeling in cell fusion on phosphatidylserine and phosphatidylserine-recognizing proteins and discuss the hypothesis that cell surface phosphatidylserine serves as a conserved "fuse me" signal regulating the time and place of cell-fusion processes.


Subject(s)
Cell Fusion , Phosphatidylserines/metabolism , Signal Transduction , Exocytosis , Humans , Virus Internalization
7.
Nat Commun ; 12(1): 495, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479215

ABSTRACT

Myomerger is a muscle-specific membrane protein involved in formation of multinucleated muscle cells by mediating the transition from the early hemifusion stage to complete fusion. Here, we considered the physical mechanism of the Myomerger action based on the hypothesis that Myomerger shifts the spontaneous curvature of the outer membrane leaflets to more positive values. We predicted, theoretically, that Myomerger generates the outer leaflet elastic stresses, which propagate into the hemifusion diaphragm and accelerate the fusion pore formation. We showed that Myomerger ectodomain indeed generates positive spontaneous curvature of lipid monolayers. We substantiated the mechanism by experiments on myoblast fusion and influenza hemagglutinin-mediated cell fusion. In both processes, the effects of Myomerger ectodomain were strikingly similar to those of lysophosphatidylcholine known to generate a positive spontaneous curvature of lipid monolayers. The control of post-hemifusion stages by shifting the spontaneous curvature of proximal membrane monolayers may be utilized in diverse fusion processes.


Subject(s)
Cell Membrane/metabolism , Membrane Fusion , Membrane Proteins/metabolism , Myoblasts/metabolism , Algorithms , Animals , Cell Fusion , Cell Line , Lipid Bilayers/metabolism , Membrane Lipids/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Theoretical , Myoblasts/cytology , NIH 3T3 Cells
8.
Arthritis Rheumatol ; 73(6): 1021-1032, 2021 06.
Article in English | MEDLINE | ID: mdl-33314777

ABSTRACT

OBJECTIVE: To identify novel heterozygous LPIN2 mutations in a patient with Majeed syndrome and characterize the pathomechanisms that lead to the development of sterile osteomyelitis. METHODS: Targeted genetic analysis and functional studies assessing monocyte responses, macrophage differentiation, and osteoclastogenesis were conducted to compare the pathogenesis of Majeed syndrome to interleukin-1 (IL-1)-mediated diseases including neonatal-onset multisystem inflammatory disease (NOMID) and deficiency of the IL-1 receptor antagonist (DIRA). RESULTS: A 4-year-old girl of mixed ethnic background presented with sterile osteomyelitis and elevated acute-phase reactants. She had a 17.8-kb deletion on the maternal LPIN2 allele and a splice site mutation, p.R517H, that variably spliced out exons 10 and 11 on the paternal LPIN2 allele. The patient achieved long-lasting remission receiving IL-1 blockade with canakinumab. Compared to controls, monocytes and monocyte-derived M1-like macrophages from the patient with Majeed syndrome and those with NOMID or DIRA had elevated caspase 1 activity and IL-1ß secretion. In contrast, lipopolysaccharide-stimulated, monocyte-derived, M2-like macrophages from the patient with Majeed syndrome released higher levels of osteoclastogenic mediators (IL-8, IL-6, tumor necrosis factor, CCL2, macrophage inflammatory protein 1α/ß, CXCL8, and CXCL1) compared to NOMID patients and healthy controls. Accelerated osteoclastogenesis in the patient with Majeed syndrome was associated with higher NFATc1 levels, enhanced JNK/MAPK, and reduced Src kinase activation, and partially responded to JNK inhibition and IL-1 (but not IL-6) blockade. CONCLUSION: We report 2 novel compound heterozygous disease-causing mutations in LPIN2 in an American patient with Majeed syndrome. LPIN2 deficiency drives differentiation of proinflammatory M2-like macrophages and enhances intrinsic osteoclastogenesis. This provides a model for the pathogenesis of sterile osteomyelitis which differentiates Majeed syndrome from other IL-1-mediated autoinflammatory diseases.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , Immunologic Deficiency Syndromes/genetics , Inflammation/genetics , Macrophages/immunology , Nuclear Proteins/genetics , Osteogenesis/genetics , Osteomyelitis/genetics , Anemia, Dyserythropoietic, Congenital/drug therapy , Anemia, Dyserythropoietic, Congenital/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Case-Control Studies , Child, Preschool , Cryopyrin-Associated Periodic Syndromes/genetics , Cryopyrin-Associated Periodic Syndromes/immunology , Female , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Heterozygote , Humans , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/immunology , Inflammation/immunology , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinases/metabolism , NFATC Transcription Factors/metabolism , Nuclear Proteins/immunology , Osteomyelitis/drug therapy , Osteomyelitis/immunology , src-Family Kinases/metabolism
9.
Bio Protoc ; 10(5): e3544, 2020 Mar 05.
Article in English | MEDLINE | ID: mdl-33659518

ABSTRACT

Lipid mixing (redistribution of lipid probes between fusing membranes) has been widely used to study early stages of relatively fast viral and intracellular fusion processes that take seconds to minutes. Lipid mixing assays are especially important for identification of hemifusion intermediates operationally defined as lipid mixing without content mixing. Due to unsynchronized character and the slow rate of the differentiation processes that prime the cells for cell-cell fusion processes in myogenesis, osteoclastogenesis and placentogenesis, these fusions take days. Application of lipid mixing assays to detect early fusion intermediates in these very slow fusion processes must consider the continuous turnover of plasma membrane components and potential fusion-unrelated exchange of the lipid probes between the membranes. Here we describe the application of lipid mixing assay in our work on myoblast fusion stage in development and regeneration of skeletal muscle cells. Our approach utilizes conventional in vitro model of myogenic differentiation and fusion based on murine C2C12 cells. When we observe the appearance of first multinucleated cells, we lift the cells and label them with either fluorescent lipid DiI as a membrane probe or CellTrackerTM Green as a content probe. Redistribution of the probes between the cells is scored by fluorescence microscopy. Hemifused cells are identified as mononucleated cells labeled with both content- and membrane probes. The interpretation must be supported by a system of negative controls with fusion-incompetent cells to account for and minimize contributions of fusion-unrelated exchange of the lipid probes. This approach with minor modifications has been used for investigating fusion of primary murine myoblasts, osteoclast precursors and fusion mediated by a gamete fusogen HAP2, and likely can be adopted for other slow cell-cell fusion processes.

10.
Sci Rep ; 9(1): 17637, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31776415

ABSTRACT

Retroviral transduction is routinely used to generate cell lines expressing exogenous non-viral genes. Here, we show that human cells transduced to stably express GFP transfer GFP gene to non-transduced cells. This horizontal gene transfer was mediated by a fraction of extracellular membrane vesicles that were released by the transduced cells. These vesicles carried endogenous retroviral envelope protein syncytin 1 and essentially acted as replication-competent retroviruses. The ability to transfer the GFP gene correlated with the levels of syncytin 1 expression in the transduced cells and depended on the fusogenic activity of this protein, substantiating the hypothesis that endogenous syncytin 1 mediates fusion stage in the delivery of extracellular vesicle cargo into target cells. Our findings suggest that testing for replication-competent retroviruses, a routine safety test for transduced cell products in clinical studies, should be also carried out for cell lines generated by retroviral vectors in in vitro studies.


Subject(s)
Gene Products, env/metabolism , Pregnancy Proteins/metabolism , Retroviridae/genetics , Transduction, Genetic/methods , Animals , Blotting, Western , Cell Line , Genetic Markers/genetics , Green Fluorescent Proteins/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction
11.
J Cell Biol ; 218(5): 1436-1451, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30936162

ABSTRACT

Cell-cell fusion remains the least understood type of membrane fusion process. However, the last few years have brought about major advances in understanding fusion between gametes, myoblasts, macrophages, trophoblasts, epithelial, cancer, and other cells in normal development and in diseases. While different cell fusion processes appear to proceed via similar membrane rearrangements, proteins that have been identified as necessary and sufficient for cell fusion (fusogens) use diverse mechanisms. Some fusions are controlled by a single fusogen; other fusions depend on several proteins that either work together throughout the fusion pathway or drive distinct stages. Furthermore, some fusions require fusogens to be present on both fusing membranes, and in other fusions, fusogens have to be on only one of the membranes. Remarkably, some of the proteins that fuse cells also sculpt single cells, repair neurons, promote scission of endocytic vesicles, and seal phagosomes. In this review, we discuss the properties and diversity of the known proteins mediating cell-cell fusion and highlight their different working mechanisms in various contexts.


Subject(s)
Cell Fusion , Cell Membrane/metabolism , Membrane Fusion , Membrane Proteins/metabolism , Animals , Humans
12.
Mol Cancer Res ; 17(3): 806-820, 2019 03.
Article in English | MEDLINE | ID: mdl-30587522

ABSTRACT

Poorly understood interactions with nonmalignant cells within the tumor microenvironment play an important role in cancer progression. Here, we explored interactions between prostate cancer and muscle cells that surround the prostate. We found that coculturing of prostate cancer cells with skeletal or smooth muscle cells expands the subpopulations of cancer cells with features characteristic of cancer stem-like cells, including anchorage-independent growth, elevated CD133 expression, and drug resistance. These changes in the properties of cancer cells depend on: (i) the muscle cell-induced increases in the concentrations of interleukins 4 and 13; (ii) the cytokine-induced upregulation of the expression of syncytin 1 and annexin A5; and (iii) cancer cell fusion. In human prostate cancer tissues, expression of syncytin 1 and annexin A5, proteins that we found to be required for the cell fusion, positively correlated with the cancer development suggesting that these proteins can be used as biomarkers to evaluate cancer progression and potential therapeutic targets. IMPLICATIONS: The discovered effects of muscle cells on prostate cancer cells reveal a novel and specific pathway by which muscle cells in the microenvironment of prostate cancer cells promote cell fusion and cancer progression.


Subject(s)
Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Male , Prostatic Neoplasms/pathology
13.
Dev Cell ; 46(6): 767-780.e7, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30197239

ABSTRACT

Classic mechanisms for membrane fusion involve transmembrane proteins that assemble into complexes and dynamically alter their conformation to bend membranes, leading to mixing of membrane lipids (hemifusion) and fusion pore formation. Myomaker and Myomerger govern myoblast fusion and muscle formation but are structurally divergent from traditional fusogenic proteins. Here, we show that Myomaker and Myomerger independently mediate distinct steps in the fusion pathway, where Myomaker is involved in membrane hemifusion and Myomerger is necessary for fusion pore formation. Mechanistically, we demonstrate that Myomerger is required on the cell surface where its ectodomains stress membranes. Moreover, we show that Myomerger drives fusion completion in a heterologous system independent of Myomaker and that a Myomaker-Myomerger physical interaction is not required for function. Collectively, our data identify a stepwise cell fusion mechanism in myoblasts where different proteins are delegated to perform unique membrane functions essential for membrane coalescence.


Subject(s)
Cell Differentiation , Membrane Fusion , Membrane Proteins/physiology , Morphogenesis , Muscle Proteins/physiology , Myoblasts/physiology , Animals , Cell Communication , Cell Fusion , Mice , Mice, Knockout , Muscle Development , Myoblasts/cytology
14.
Sci Rep ; 8(1): 1768, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29379067

ABSTRACT

Cell-cell fusion is a key stage in development and maintenance of multinucleated cells that resorb bones and form our skeletal muscles and placenta. Here, we focus on osteoclast formation to suggest new ways of unbiased presentation of cell fusion at given conditions that combine empirical cumulative distribution function for the sizes of multinucleated cells with the total number of cell-cell fusion events, which generate these cells.


Subject(s)
Osteoclasts/physiology , Animals , Bone Resorption/physiopathology , Bone and Bones/physiology , Cell Differentiation/physiology , Cell Fusion/methods , Cells, Cultured , Humans , Mice , RAW 264.7 Cells
15.
J Biol Chem ; 293(1): 254-270, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29101233

ABSTRACT

Bone-resorbing multinucleated osteoclasts that play a central role in the maintenance and repair of our bones are formed from bone marrow myeloid progenitor cells by a complex differentiation process that culminates in fusion of mononuclear osteoclast precursors. In this study, we uncoupled the cell fusion step from both pre-fusion stages of osteoclastogenic differentiation and the post-fusion expansion of the nascent fusion connections. We accumulated ready-to-fuse cells in the presence of the fusion inhibitor lysophosphatidylcholine and then removed the inhibitor to study synchronized cell fusion. We found that osteoclast fusion required the dendrocyte-expressed seven transmembrane protein (DC-STAMP)-dependent non-apoptotic exposure of phosphatidylserine at the surface of fusion-committed cells. Fusion also depended on extracellular annexins, phosphatidylserine-binding proteins, which, along with annexin-binding protein S100A4, regulated fusogenic activity of syncytin 1. Thus, in contrast to fusion processes mediated by a single protein, such as epithelial cell fusion in Caenorhabditis elegans, the cell fusion step in osteoclastogenesis is controlled by phosphatidylserine-regulated activity of several proteins.


Subject(s)
Gene Products, env/metabolism , Osteogenesis/physiology , Phosphatidylserines/physiology , Pregnancy Proteins/metabolism , Animals , Annexins/metabolism , Bone Resorption/metabolism , Bone and Bones/metabolism , Cell Differentiation , Cell Fusion/methods , Cell Line , Cell Membrane/metabolism , Gene Products, env/physiology , Hematopoiesis , Humans , Membrane Fusion/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Osteoclasts/physiology , Phosphatidylserines/metabolism , Pregnancy Proteins/physiology , S100 Calcium-Binding Protein A4/metabolism
16.
J Biol Chem ; 292(42): 17272-17289, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28860190

ABSTRACT

Multinucleated skeletal muscle fibers form through the fusion of myoblasts during development and regeneration. Previous studies identified myomaker (Tmem8c) as a muscle-specific membrane protein essential for fusion. However, the specific function of myomaker and how its function is regulated are unknown. To explore these questions, we first examined the cellular localization of endogenous myomaker. Two independent antibodies showed that whereas myomaker does localize to the plasma membrane in cultured myoblasts, the protein also resides in the Golgi and post-Golgi vesicles. These results raised questions regarding the precise cellular location of myomaker function and mechanisms that govern myomaker trafficking between these cellular compartments. Using a synchronized fusion assay, we demonstrated that myomaker functions at the plasma membrane to drive fusion. Trafficking of myomaker is regulated by palmitoylation of C-terminal cysteine residues that allows Golgi localization. Moreover, dissection of the C terminus revealed that palmitoylation was not sufficient for complete fusogenic activity suggesting a function for other amino acids within this C-terminal region. Indeed, C-terminal mutagenesis analysis highlighted the importance of a C-terminal leucine for function. These data reveal that myoblast fusion requires myomaker activity at the plasma membrane and is potentially regulated by proper myomaker trafficking.


Subject(s)
Antigens, Differentiation/metabolism , Golgi Apparatus/metabolism , Lipoylation/physiology , Membrane Fusion/physiology , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Myoblasts, Skeletal/metabolism , Animals , Antigens, Differentiation/genetics , Cell Line , Golgi Apparatus/genetics , Membrane Proteins/genetics , Mice , Muscle Proteins/genetics , Myoblasts, Skeletal/cytology , Protein Domains , Protein Transport/physiology
17.
Cell Host Microbe ; 22(1): 99-110.e7, 2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28704658

ABSTRACT

HIV-1 entry into host cells starts with interactions between the viral envelope glycoprotein (Env) and cellular CD4 receptors and coreceptors. Previous work has suggested that efficient HIV entry also depends on intracellular signaling, but this remains controversial. Here we report that formation of the pre-fusion Env-CD4-coreceptor complexes triggers non-apoptotic cell surface exposure of the membrane lipid phosphatidylserine (PS). HIV-1-induced PS redistribution depends on Ca2+ signaling triggered by Env-coreceptor interactions and involves the lipid scramblase TMEM16F. Externalized PS strongly promotes Env-mediated membrane fusion and HIV-1 infection. Blocking externalized PS or suppressing TMEM16F inhibited Env-mediated fusion. Exogenously added PS promoted fusion, with fusion dependence on PS being especially strong for cells with low surface density of coreceptors. These findings suggest that cell-surface PS acts as an important cofactor that promotes the fusogenic restructuring of pre-fusion complexes and likely focuses the infection on cells conducive to PS signaling.


Subject(s)
HIV Infections/virology , HIV-1/physiology , HIV-1/pathogenicity , Membrane Fusion/physiology , Phosphatidylserines/metabolism , Virus Activation/physiology , Virus Internalization , Amides/antagonists & inhibitors , Anoctamins/metabolism , Antibodies, Monoclonal , Benzylamines , CD4 Antigens/metabolism , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cyclams , HEK293 Cells , HeLa Cells , Heterocyclic Compounds/antagonists & inhibitors , Host-Pathogen Interactions/physiology , Humans , Phospholipid Transfer Proteins/metabolism , Quaternary Ammonium Compounds/antagonists & inhibitors , Receptors, CCR5/drug effects , Receptors, CCR5/immunology , Receptors, CXCR4/drug effects , Signal Transduction , Viral Envelope Proteins/metabolism , Virus Attachment , Virus Replication/physiology
18.
J Cell Biol ; 216(3): 571-581, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28137780

ABSTRACT

Cell-cell fusion is inherent to sexual reproduction. Loss of HAPLESS 2/GENERATIVE CELL SPECIFIC 1 (HAP2/GCS1) proteins results in gamete fusion failure in diverse organisms, but their exact role is unclear. In this study, we show that Arabidopsis thaliana HAP2/GCS1 is sufficient to promote mammalian cell-cell fusion. Hemifusion and complete fusion depend on HAP2/GCS1 presence in both fusing cells. Furthermore, expression of HAP2 on the surface of pseudotyped vesicular stomatitis virus results in homotypic virus-cell fusion. We demonstrate that the Caenorhabditis elegans Epithelial Fusion Failure 1 (EFF-1) somatic cell fusogen can replace HAP2/GCS1 in one of the fusing membranes, indicating that HAP2/GCS1 and EFF-1 share a similar fusion mechanism. Structural modeling of the HAP2/GCS1 protein family predicts that they are homologous to EFF-1 and viral class II fusion proteins (e.g., Zika virus). We name this superfamily Fusexins: fusion proteins essential for sexual reproduction and exoplasmic merger of plasma membranes. We suggest a common origin and evolution of sexual reproduction, enveloped virus entry into cells, and somatic cell fusion.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Carrier Proteins/metabolism , Germ Cells/metabolism , Viral Fusion Proteins/metabolism , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Fusion/methods , Cell Line , Cell Membrane/metabolism , Cell Membrane/physiology , Cricetinae , Membrane Fusion/physiology , Membrane Glycoproteins/metabolism
20.
Virology ; 488: 20-7, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26590794

ABSTRACT

High-throughput techniques are needed to analyze individual virions to understand how viral heterogeneity translates into pathogenesis since in bulk analysis the individual characteristics of virions are lost. Individual Dengue virions (DENV) undergo a maturation that involves a proteolytic cleavage of prM precursor into virion-associated M protein. Here, using a new nanoparticle-based technology, "flow virometry", we compared the maturation of individual DENV produced by BHK-21 and LoVo cells. The latter lacks the furin-protease that mediates prM cleavage. We found that prM is present on about 50% of DENV particles produced in BHK-21 cells and about 85% of DENV virions produced in LoVo, indicating an increase in the fraction of not fully matured virions. Flow virometry allows us to quantify the number of fully mature particles in DENV preparations and proves to be a useful method for studying heterogeneity of the surface proteins of various viruses.


Subject(s)
Dengue Virus/physiology , Viral Envelope Proteins/metabolism , Virion/metabolism , Virology/methods , Animals , Cell Line , Cricetinae , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...