Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
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.
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
3.
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
4.
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
5.
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
6.
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.

7.
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
8.
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
9.
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
10.
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
11.
Sci Rep ; 5: 18246, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26667898

ABSTRACT

Repair and regeneration of the injured skeletal myofiber involves fusion of intracellular vesicles with sarcolemma and fusion of the muscle progenitor cells respectively. In vitro experiments have identified involvement of Annexin A1 (Anx A1) in both these fusion processes. To determine if Anx A1 contributes to these processes during muscle repair in vivo, we have assessed muscle growth and repair in Anx A1-deficient mouse (AnxA1-/-). We found that the lack of Anx A1 does not affect the muscle size and repair of myofibers following focal sarcolemmal injury and lengthening contraction injury. However, the lack of Anx A1 delayed muscle regeneration after notexin-induced injury. This delay in muscle regeneration was not caused by a slowdown in proliferation and differentiation of satellite cells. Instead, lack of Anx A1 lowered the proportion of differentiating myoblasts that managed to fuse with the injured myofibers by days 5 and 7 after notexin injury as compared to the wild type (w.t.) mice. Despite this early slowdown in fusion of Anx A1-/- myoblasts, regeneration caught up at later times post injury. These results establish in vivo role of Anx A1 in cell fusion required for myofiber regeneration and not in intracellular vesicle fusion needed for repair of myofiber sarcolemma.


Subject(s)
Annexin A1/deficiency , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiology , Wound Healing/genetics , Animals , Cell Fusion , Female , Male , Mice , Mice, Knockout , Muscle Contraction/genetics , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Sarcolemma/metabolism , Sarcolemma/ultrastructure
12.
Prostate ; 75(9): 907-16, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25728608

ABSTRACT

BACKGROUND: PTEN/AKT signaling plays a key role in prostate cancer development and maintenance of prostate cancer stem cells. How other oncogenes or tumor suppressors interact with this pathway remain to be elucidated. SLUG is an zinc finger transcription factor of the Snail superfamily, and it promotes cancer metastasis and determines the mammary stem cell state. METHODS: SLUG was overexpressed in cells by retroviral vector and knockdown of SLUG and PTEN was mediated by shRNAs-expressing lentiviruses. Expression level of SLUG and PTEN was examined by Western blot, RT-PCR, and qPCR analyses. PTEN promoter activity was measured by luciferase reporter assay. ChIP assay was used to measure the binding between SLUG and the PTEN promoter in vivo. RESULT: We showed that overexpression of SLUG decreased expression of PTEN tumor repressor in prostate cancer cell lines 22RV1 and DU145; conversely, knockdown of SLUG expression elevated PTEN expresson at both protein and RNA level in these cells. We demonstrated that SLUG overexpression inhibits PTEN promoter activity through the proximal promoter region in prostate cancer cells. By ChIP assay, we confirmed that SLUG directly binds to the PTEN promoter region covering the E-box sites. We also showed that Slug deficiency leads to an increased expression of PTEN in mouse embryo fibroblasts and prostate tissues. Importantly, we found that overexpression of SLUG increases drug resistance of DU145 prostate cancer cell line and knockdown of SLUG by shRNA sensitizes DU145 cell line to chemotherapeutic drugs. We further demonstrated that PTEN knockdown converts drug sensitivity of DU145 cells expressing SLUG shRNA to anticancer drugs. CONCLUSION: We provide compelling evidence showing that PTEN is a direct functional target of SLUG. Our findings offer new insight in the regulation of the PTEN/AKT pathway and provide a molecular basis for potential targeted therapies of prostate cancer Prostate 75:907-916, 2015. © 2015 Wiley Periodicals, Inc.


Subject(s)
PTEN Phosphohydrolase/biosynthesis , Prostatic Neoplasms/metabolism , Transcription Factors/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/genetics
13.
Biochem J ; 464(3): 293-300, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25336256

ABSTRACT

Macrophage fusion that leads to osteoclast formation is one of the most important examples of cell-cell fusion in development, tissue homoeostasis and immune response. Protein machinery that fuses macrophages remains to be identified. In the present study, we explored the fusion stage of osteoclast formation for RAW macrophage-like murine cells and for macrophages derived from human monocytes. To uncouple fusion from the preceding differentiation processes, we accumulated fusion-committed cells in the presence of LPC (lysophosphatidylcholine) that reversibly blocks membrane merger. After 16 h, we removed LPC and observed cell fusion events that would normally develop within 16 h develop instead within 30-90 min. Thus, whereas osteoclastogenesis, generally, takes several days, our approach allowed us to focus on an hour in which we observe robust fusion between the cells. Complementing syncytium formation assay with a novel membrane merger assay let us study the synchronized fusion events downstream of a local merger between two plasma membranes, but before expansion of nascent membrane connections and complete unification of the cells. We found that the expansion of membrane connections detected as a growth of multinucleated osteoclasts depends on dynamin activity. In contrast, a merger between the plasma membranes of the two cells was not affected by inhibitors of dynamin GTPase. Thus dynamin that was recently found to control late stages of myoblast fusion also controls late stages of macrophage fusion, revealing an intriguing conserved mechanistic motif shared by diverse cell-cell fusion processes.


Subject(s)
Dynamin II/metabolism , Macrophages/physiology , Osteoclasts/physiology , Animals , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Fusion , Cells, Cultured , Dynamin II/genetics , Humans , Lysophosphatidylcholines/pharmacology , Macrophages/drug effects , Mice , Osteoclasts/drug effects , RNA, Small Interfering/genetics
14.
Hum Mol Genet ; 23(18): 4745-57, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24760765

ABSTRACT

While spinal muscular atrophy (SMA) is characterized by motor neuron degeneration, it is unclear whether and how much survival motor neuron (SMN) protein deficiency in muscle contributes to the pathophysiology of the disease. There is increasing evidence from patients and SMA model organisms that SMN deficiency causes intrinsic muscle defects. Here we investigated the role of SMN in muscle development using muscle cell lines and primary myoblasts. Formation of multinucleate myotubes by SMN-deficient muscle cells is inhibited at a stage preceding plasma membrane fusion. We found increased expression and reduced induction of key muscle development factors, such as MyoD and myogenin, with differentiation of SMN-deficient cells. In addition, SMN-deficient muscle cells had impaired cell migration and altered organization of focal adhesions and the actin cytoskeleton. Partially restoring SMN inhibited the premature expression of muscle differentiation markers, corrected the cytoskeletal abnormalities and improved myoblast fusion. These findings are consistent with a role for SMN in myotube formation through effects on muscle differentiation and cell motility.


Subject(s)
Focal Adhesions/metabolism , Muscle Development , Muscle Fibers, Skeletal/metabolism , Myogenin/genetics , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cell Differentiation , Cell Line , Cell Movement , Gene Expression Regulation , Humans , Mice , Muscle Fibers, Skeletal/cytology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenin/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Survival of Motor Neuron 1 Protein/genetics
15.
J Cell Biol ; 200(1): 109-23, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23277424

ABSTRACT

Myoblast fusion into multinucleated myotubes is a crucial step in skeletal muscle development and regeneration. Here, we accumulated murine myoblasts at the ready-to-fuse stage by blocking formation of early fusion intermediates with lysophosphatidylcholine. Lifting the block allowed us to explore a largely synchronized fusion. We found that initial merger of two cell membranes detected as lipid mixing involved extracellular annexins A1 and A5 acting in a functionally redundant manner. Subsequent stages of myoblast fusion depended on dynamin activity, phosphatidylinositol(4,5)bisphosphate content, and cell metabolism. Uncoupling fusion from preceding stages of myogenesis will help in the analysis of the interplay between protein machines that initiate and complete cell unification and in the identification of additional protein players controlling different fusion stages.


Subject(s)
Annexin A1/metabolism , Annexin A5/metabolism , Cell Membrane/metabolism , Dynamins/metabolism , Muscle Development/physiology , Myoblasts/metabolism , Animals , Annexin A1/genetics , Annexin A5/genetics , Cell Fusion , Cell Line , Cell Membrane/genetics , Dynamins/genetics , Mice , Mice, Knockout , Myoblasts/cytology , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism
16.
Biochem J ; 440(2): 185-93, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21895608

ABSTRACT

Cell-to-cell fusion plays an important role in normal physiology and in different pathological conditions. Early fusion stages mediated by specialized proteins and yielding fusion pores are followed by a pore expansion stage that is dependent on cell metabolism and yet unidentified machinery. Because of a similarity of membrane bending in the fusion pore rim and in highly curved intracellular membrane compartments, in the present study we explored whether changes in the activity of the proteins that generate these compartments affect cell fusion initiated by protein fusogens of influenza virus and baculovirus. We raised the intracellular concentration of curvature-generating proteins in cells by either expressing or microinjecting the ENTH (epsin N-terminal homology) domain of epsin or by expressing the GRAF1 (GTPase regulator associated with focal adhesion kinase 1) BAR (Bin/amphiphysin/Rvs) domain or the FCHo2 (FCH domain-only protein 2) F-BAR domain. Each of these treatments promoted syncytium formation. Cell fusion extents were also influenced by treatments targeting the function of another curvature-generating protein, dynamin. Cell-membrane-permeant inhibitors of dynamin GTPase blocked expansion of fusion pores and dominant-negative mutants of dynamin influenced the syncytium formation extents. We also report that syncytium formation is inhibited by reagents lowering the content and accessibility of PtdIns(4,5)P(2), an important regulator of intracellular membrane remodelling. Our findings indicate that fusion pore expansion at late stages of cell-to-cell fusion is mediated, directly or indirectly, by intracellular membrane-shaping proteins.


Subject(s)
Cell Fusion , Cell Membrane/ultrastructure , Membrane Fusion/physiology , Membrane Proteins/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Dynamins/metabolism , Giant Cells/physiology , Hemagglutinins, Viral/physiology , Mice , NIH 3T3 Cells , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Structure, Tertiary
17.
Curr Biol ; 20(12): 1117-21, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20537541

ABSTRACT

The apicomplexan parasite Plasmodium falciparum causes malignant malaria. The mechanism of parasite egress from infected erythrocytes that disseminate parasites in the host at the end of each asexual cycle is unknown. Two new stages of the egress program are revealed: (1) swelling of the parasitophorous vacuole accompanied by shrinkage of the erythrocyte compartment, and (2) poration of the host cell membrane seconds before erythrocyte rupture because of egress. Egress was inhibited in dehydrated cells from patients with sickle cell disease in accord with experimental dehydration of normal cells, suggesting that vacuole swelling involves intake of water from the erythrocyte compartment. Erythrocyte membrane poration occurs in relaxed cells, thus excluding involvement of osmotic pressure in this process. Poration does not depend on cysteine protease activity, because protease inhibition blocks egress but not poration, and poration is required for the parasite cycle because the membrane sealant P1107 interferes with egress. We suggest the following egress program: parasites initiate water influx into the vacuole from the erythrocyte cytosol to expand the vacuole for parasite separation and vacuole rupture upon its critical swelling. Separated parasites leave the erythrocyte by breaching its membrane, weakened by putative digestion of erythrocyte cytoskeleton and membrane poration.


Subject(s)
Anemia, Sickle Cell/blood , Erythrocytes, Abnormal/parasitology , Erythrocytes/parasitology , Plasmodium falciparum/physiology , Animals , Base Sequence , Cell Membrane , DNA Primers , Humans
18.
Dev Cell ; 12(5): 683-98, 2007 May.
Article in English | MEDLINE | ID: mdl-17488621

ABSTRACT

Cell fusion is fundamental for reproduction and organ formation. Fusion between most C. elegans epithelial cells is mediated by the EFF-1 fusogen. However, fusion between the anchor cell and the utse syncytium that establishes a continuous uterine-vulval tube proceeds normally in eff-1 mutants. By isolating mutants where the anchor-cell fails to fuse, we identified aff-1. AFF-1 ectopic expression results in fusion of cells that normally do not fuse in C. elegans. The fusogen activity of AFF-1 was further confirmed by its ability to fuse heterologous cells. AFF-1 and EFF-1 differ in their fusogenic activity and expression patterns but share eight conserved predicted disulfide bonds in their ectodomains, including a putative TGF-beta-type-I-Receptor domain. We found that FOS-1, the Fos transcription factor ortholog that controls anchor-cell invasion during nematode development, is a specific activator of aff-1-mediated anchor-cell fusion. Thus, FOS-1 links cell invasion and fusion in a developmental cascade.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/chemistry , Cell Fusion , Cytoplasm/metabolism , Embryo, Nonmammalian/cytology , Epithelial Cells/cytology , Female , Insecta/cytology , Models, Biological , Molecular Sequence Data , Mutation/genetics , Phenotype , Proto-Oncogene Proteins c-fos/chemistry , Transcription Factors/chemistry , Vulva/cytology , Vulva/growth & development
19.
Dev Cell ; 11(4): 471-81, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17011487

ABSTRACT

During cell-cell fusion, two cells' plasma membranes merge, allowing the cytoplasms to mix and form a syncytium. Little is known about the mechanisms of cell fusion. Here, we asked whether eff-1, shown previously to be essential for fusion in Caenorhabditis elegans, acts directly in the fusion machinery. We show that expression of EFF-1 transmembrane protein drives fusion of heterologous cells into multinucleate syncytia. We obtained evidence that EFF-1-mediated fusion involves a hemifusion intermediate characterized by membrane mixing without cytoplasm mixing. Furthermore, syncytiogenesis requires EFF-1 in both fusing cells. To test whether this mechanism also applies in vivo, we conducted genetic mosaic analysis of C. elegans and found that homotypic epidermal fusion requires EFF-1 in both cells. Thus, although EFF-1-mediated fusion shares characteristics with viral and intracellular fusion, including an apparent hemifusion step, it differs from these reactions in the homotypic organization of the fusion machinery.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Cell Membrane/physiology , Membrane Glycoproteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Fusion , Cell Membrane/metabolism , Cells, Cultured , Embryo, Nonmammalian , Genes, Reporter , Green Fluorescent Proteins/metabolism , Kinetics , Membrane Glycoproteins/genetics , Models, Biological , Mosaicism , Spodoptera/cytology , Time Factors , Transfection , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...