Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 633
Filter
Add more filters

Publication year range
1.
Cell ; 184(12): 3192-3204.e16, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33974910

ABSTRACT

Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is initiated by binding of the viral Spike protein to host receptor angiotensin-converting enzyme 2 (ACE2), followed by fusion of viral and host membranes. Although antibodies that block this interaction are in emergency use as early coronavirus disease 2019 (COVID-19) therapies, the precise determinants of neutralization potency remain unknown. We discovered a series of antibodies that potently block ACE2 binding but exhibit divergent neutralization efficacy against the live virus. Strikingly, these neutralizing antibodies can inhibit or enhance Spike-mediated membrane fusion and formation of syncytia, which are associated with chronic tissue damage in individuals with COVID-19. As revealed by cryoelectron microscopy, multiple structures of Spike-antibody complexes have distinct binding modes that not only block ACE2 binding but also alter the Spike protein conformational cycle triggered by ACE2 binding. We show that stabilization of different Spike conformations leads to modulation of Spike-mediated membrane fusion with profound implications for COVID-19 pathology and immunity.


Subject(s)
Antibodies, Neutralizing/chemistry , Giant Cells/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/metabolism , Binding Sites , CHO Cells , COVID-19/pathology , COVID-19/virology , Cricetinae , Cricetulus , Cryoelectron Microscopy , Giant Cells/cytology , Humans , Membrane Fusion , Peptide Library , Protein Binding , Protein Domains , Protein Structure, Quaternary , SARS-CoV-2/isolation & purification , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism
2.
Nature ; 622(7983): 562-573, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37673118

ABSTRACT

The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation1. Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2. Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3. Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4. Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development.


Subject(s)
Embryo Implantation , Embryo, Mammalian , Embryonic Development , Human Embryonic Stem Cells , Humans , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Fertilization , Gastrulation , Germ Layers/cytology , Germ Layers/embryology , Human Embryonic Stem Cells/cytology , Trophoblasts/cytology , Yolk Sac/cytology , Yolk Sac/embryology , Giant Cells/cytology
3.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-35001104

ABSTRACT

Biological systems are highly complex, yet notably ordered structures can emerge. During syncytial stage development of the Drosophila melanogaster embryo, nuclei synchronously divide for nine cycles within a single cell, after which most of the nuclei reach the cell cortex. The arrival of nuclei at the cortex occurs with remarkable positional order, which is important for subsequent cellularisation and morphological transformations. Yet, the mechanical principles underlying this lattice-like positional order of nuclei remain untested. Here, using quantification of nuclei position and division orientation together with embryo explants, we show that short-ranged repulsive interactions between microtubule asters ensure the regular distribution and maintenance of nuclear positions in the embryo. Such ordered nuclear positioning still occurs with the loss of actin caps and even the loss of the nuclei themselves; the asters can self-organise with similar distribution to nuclei in the wild-type embryo. The explant assay enabled us to deduce the nature of the mechanical interaction between pairs of nuclei. We used this to predict how the nuclear division axis orientation changes upon nucleus removal from the embryo cortex, which we confirmed in vivo with laser ablation. Overall, we show that short-ranged microtubule-mediated repulsive interactions between asters are important for ordering in the early Drosophila embryo and minimising positional irregularity.


Subject(s)
Blastoderm/metabolism , Cell Nucleus Division , Giant Cells/metabolism , Animals , Blastoderm/cytology , Cell Nucleus/metabolism , Drosophila melanogaster , Giant Cells/cytology , Microtubules/metabolism , Stress, Mechanical
4.
Development ; 146(1)2019 01 02.
Article in English | MEDLINE | ID: mdl-30509968

ABSTRACT

In teleost fish, the multinucleate yolk syncytial layer functions as an extra-embryonic signaling center to pattern mesendoderm, coordinate morphogenesis and supply nutrients to the embryo. External yolk syncytial nuclei (e-YSN) undergo microtubule-dependent movements that distribute the nuclei over the large yolk mass. How e-YSN migration proceeds, and the role of the yolk microtubules, is not understood, but it is proposed that e-YSN are pulled vegetally as the microtubule network shortens from the vegetal pole. Live imaging revealed that nuclei migrate along microtubules, consistent with a cargo model in which e-YSN are moved down the microtubules by direct association with motor proteins. We found that blocking the plus-end directed microtubule motor kinesin significantly attenuated yolk nuclear movement. Blocking the outer nuclear membrane LINC complex protein Syne2a also slowed e-YSN movement. We propose that e-YSN movement is mediated by the LINC complex, which functions as the adaptor between yolk nuclei and motor proteins. Our work provides new insights into the role of microtubules in morphogenesis of an extra-embryonic tissue and further contributes to the understanding of nuclear migration mechanisms during development.


Subject(s)
Cell Movement , Cell Nucleus/metabolism , Giant Cells/cytology , Models, Biological , Zebrafish/embryology , Zebrafish/metabolism , Animals , Dyneins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Kinesins/metabolism , Microtubules/metabolism , Time-Lapse Imaging
5.
PLoS Comput Biol ; 17(8): e1008828, 2021 08.
Article in English | MEDLINE | ID: mdl-34339411

ABSTRACT

Multinucleate cells occur in every biosphere and across the kingdoms of life, including in the human body as muscle cells and bone-forming cells. Data from filamentous fungi suggest that, even when bathed in a common cytoplasm, nuclei are capable of autonomous behaviors, including division. How does this potential for autonomy affect the organization of cellular processes between nuclei? Here we analyze a simplified model of circadian rhythm, a form of cellular oscillator, in a mathematical model of the filamentous fungus Neurospora crassa. Our results highlight a potential role played by mRNA-protein phase separation to keep mRNAs close to the nuclei from which they originate, while allowing proteins to diffuse freely between nuclei. Our modeling shows that syncytism allows for extreme mRNA efficiency-we demonstrate assembly of a robust oscillator with a transcription rate a thousand-fold less than in comparable uninucleate cells. We also show self-organized division of the labor of mRNA production, with one nucleus in a two-nucleus syncytium producing at least twice as many mRNAs as the other in 30% of cycles. This division can occur spontaneously, but division of labor can also be controlled by regulating the amount of cytoplasmic volume available to each nucleus. Taken together, our results show the intriguing richness and potential for emergent organization among nuclei in multinucleate cells. They also highlight the role of previously studied mechanisms of cellular organization, including nuclear space control and localization of mRNAs through RNA-protein phase separation, in regulating nuclear coordination.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Algorithms , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Computational Biology , Computer Simulation , Cytoplasm/genetics , Cytoplasm/metabolism , Giant Cells/cytology , Giant Cells/metabolism , Humans , Models, Genetic , Neurospora crassa/cytology , Neurospora crassa/genetics , Neurospora crassa/physiology , RNA, Fungal/genetics , RNA, Fungal/metabolism , Stochastic Processes , Transcription, Genetic
6.
Cell Mol Life Sci ; 78(17-18): 6087-6104, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34296319

ABSTRACT

Different types of multinucleated giant cells (MGCs) of myeloid origin have been described; osteoclasts are the most extensively studied because of their importance in bone homeostasis. MGCs are formed by cell-to-cell fusion, and most types have been observed in pathological conditions, especially in infectious and non-infectious chronic inflammatory contexts. The precise role of the different MGCs and the mechanisms that govern their formation remain poorly understood, likely due to their heterogeneity. First, we will introduce the main populations of MGCs derived from the monocyte/macrophage lineage. We will then discuss the known molecular actors mediating the early stages of fusion, focusing on cell-surface receptors involved in the cell-to-cell adhesion steps that ultimately lead to multinucleation. Given that cell-to-cell fusion is a complex and well-coordinated process, we will also describe what is currently known about the evolution of F-actin-based structures involved in macrophage fusion, i.e., podosomes, zipper-like structures, and tunneling nanotubes (TNT). Finally, the localization and potential role of the key fusion mediators related to the formation of these F-actin structures will be discussed. This review intends to present the current status of knowledge of the molecular and cellular mechanisms supporting multinucleation of myeloid cells, highlighting the gaps still existing, and contributing to the proposition of potential disease-specific MGC markers and/or therapeutic targets.


Subject(s)
Cell Adhesion , Giant Cells/metabolism , Myeloid Cells/metabolism , Podosomes/metabolism , Giant Cells/cytology , Humans , Integrins/metabolism , Macrophages/cytology , Macrophages/metabolism , Myeloid Cells/cytology , Myeloid Cells/ultrastructure , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis , Receptors, Immunologic/metabolism
7.
Int J Mol Sci ; 23(3)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35163532

ABSTRACT

Since the beginning of the HIV epidemic, lasting more than 30 years, the main goal of scientists was to develop effective methods for the prevention and treatment of HIV infection. Modern medicines have reduced the death rate from AIDS by 80%. However, they still have side effects and are very expensive, dictating the need to search for new drugs. Earlier, it was shown that phospholipases A2 (PLA2s) from bee and snake venoms block HIV replication, the effect being independent on catalytic PLA2 activity. However, the antiviral activity of human PLA2s against Lentiviruses depended on catalytic function and was mediated through the destruction of the viral membrane. To clarify the role of phospholipolytic activity in antiviral effects, we analyzed the anti-HIV activity of several snake PLA2s and found that the mechanisms of their antiviral activity were similar to that of mammalian PLA2. Our results indicate that snake PLA2s are capable of inhibiting syncytium formation between chronically HIV-infected cells and healthy CD4-positive cells and block HIV binding to cells. However, only dimeric PLA2s had pronounced virucidal and anti-HIV activity, which depended on their catalytic activity. The ability of snake PLA2s to inactivate the virus may provide an additional barrier to HIV infection. Thus, snake PLA2s might be considered as candidates for lead molecules in anti-HIV drug development.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/cytology , Giant Cells/cytology , HIV-1/physiology , Phospholipases A2/pharmacology , Snake Venoms/enzymology , Snakes/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Giant Cells/drug effects , Giant Cells/virology , HIV-1/drug effects , Humans , Inhibitory Concentration 50 , Reptilian Proteins/pharmacology , Snakes/classification , Virus Activation/drug effects , Virus Attachment/drug effects
8.
Cytokine ; 142: 155486, 2021 06.
Article in English | MEDLINE | ID: mdl-33721618

ABSTRACT

Truncated tryptophanyl-tRNA synthetase (mini-TrpRS), like any other aminoacyl-tRNA synthetases, canonically functions as a protein synthesis enzyme. Here we provide evidence for an additional signaling role of mini-TrpRS in the formation of monocyte-derived multinuclear giant cells (MGCs). Interferon-gamma (IFNγ) readily induced monocyte aggregation leading to MGC formation with paralleled marked upregulation of mini-TrpRS. Small interfering (si)RNA, targeting mini-TrpRS in the presence of IFNγ prevented monocyte aggregation. Moreover, blockade of mini-TrpRS, either by siRNA, or the cognate amino acid and decoy substrate D-Tryptophan to prevent mini-TrpRS signaling, resulted in a marked reduction in expression of the purinergic receptor P2X 7 (P2RX7) in monocytes activated by IFNγ. Our findings identify mini-TrpRS as a critical signaling molecule in a mechanism by which IFNγ initiates monocyte-derived giant cell formation.


Subject(s)
Giant Cells/cytology , Giant Cells/enzymology , Interferon-gamma/pharmacology , Monocytes/cytology , Tryptophan-tRNA Ligase/metabolism , Cell Aggregation/drug effects , Down-Regulation/drug effects , Giant Cells/drug effects , Humans , Models, Biological , Receptors, Purinergic/metabolism , Signal Transduction/drug effects , THP-1 Cells , Up-Regulation/drug effects
9.
PLoS Genet ; 14(1): e1007174, 2018 01.
Article in English | MEDLINE | ID: mdl-29337989

ABSTRACT

Despite extensive work on the mechanisms that generate plasma membrane furrows, understanding how cells are able to dynamically regulate furrow dimensions is an unresolved question. Here, we present an in-depth characterization of furrow behaviors and their regulation in vivo during early Drosophila morphogenesis. We show that the deepening in furrow dimensions with successive nuclear cycles is largely due to the introduction of a new, rapid ingression phase (Ingression II). Blocking the midblastula transition (MBT) by suppressing zygotic transcription through pharmacological or genetic means causes the absence of Ingression II, and consequently reduces furrow dimensions. The analysis of compound chromosomes that produce chromosomal aneuploidies suggests that multiple loci on the X, II, and III chromosomes contribute to the production of differentially-dimensioned furrows, and we track the X-chromosomal contribution to furrow lengthening to the nullo gene product. We further show that checkpoint proteins are required for furrow lengthening; however, mitotic phases of the cell cycle are not strictly deterministic for furrow dimensions, as a decoupling of mitotic phases with periods of active ingression occurs as syncytial furrow cycles progress. Finally, we examined the turnover of maternal gene products and find that this is a minor contributor to the developmental regulation of furrow morphologies. Our results suggest that cellularization dynamics during cycle 14 are a continuation of dynamics established during the syncytial cycles and provide a more nuanced view of developmental- and MBT-driven morphogenesis.


Subject(s)
Blastula/cytology , Blastula/embryology , Cell Division , Cell Membrane , Morphogenesis/genetics , Zygote/physiology , Animals , Animals, Genetically Modified , Cell Division/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Embryonic Development/physiology , Female , Gene Expression Regulation, Developmental , Giant Cells/cytology , Giant Cells/metabolism , Giant Cells/ultrastructure , Male , Zygote/metabolism
10.
Bull Exp Biol Med ; 171(4): 508-512, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34542766

ABSTRACT

In the era of molecular biology and atomic force microscopy, some important macroscopic issues such as simultaneous bidirectional axonal flow or neuronal multinucleosis remain unaddressed. However, these issues have to be addressed, because they distort the results of our current achievements. Using videorecording technique, we studied adhesive contacts between neurons and their processes and kinetics of anastomosis retraction between the cell bodies up to their complete fusion with introduction of neurites into the cell cytoplasm and formation of binuclear cells. Three proofs refuting the mechanism of binuclearity formation by amitosis are presented. Live trinuclear neurons without signs of amitotic division were identified. Electron microscopy showed that fusion of many living neurons into one simplest during centrifugation of isolated cells.


Subject(s)
Giant Cells/cytology , Neurons/physiology , Animals , Cell Fusion , Cell Nucleus/ultrastructure , Cells, Cultured , Giant Cells/physiology , Giant Cells/ultrastructure , Kinetics , Lymnaea , Microscopy, Electron , Neurons/cytology , Neurons/ultrastructure
11.
Pflugers Arch ; 472(5): 561-570, 2020 05.
Article in English | MEDLINE | ID: mdl-32415460

ABSTRACT

We previously demonstrated that a two-cell syncytium, composed of a ventricular myocyte and an mHCN2 expressing cell, recapitulated most properties of in vivo biological pacing induced by mHCN2-transfected hMSCs in the canine ventricle. Here, we use the two-cell syncytium, employing dynamic clamp, to study the roles of gf (pacemaker conductance), gK1 (background K+ conductance), and gj (intercellular coupling conductance) in biological pacing. We studied gf and gK1 in single HEK293 cells expressing cardiac sodium current channel Nav1.5 (SCN5A). At fixed gf, increasing gK1 hyperpolarized the cell and initiated pacing. As gK1 increased, rate increased, then decreased, finally ceasing at membrane potentials near EK. At fixed gK1, increasing gf depolarized the cell and initiated pacing. With increasing gf, rate increased reaching a plateau, then decreased, ceasing at a depolarized membrane potential. We studied gj via virtual coupling with two non-adjacent cells, a driver (HEK293 cell) in which gK1 and gf were injected without SCN5A and a follower (HEK293 cell), expressing SCN5A. At the chosen values of gK1 and gf oscillations initiated in the driver, when gj was increased synchronized pacing began, which then decreased by about 35% as gj approached 20 nS. Virtual uncoupling yielded similar insights into gj. We also studied subthreshold oscillations in physically and virtually coupled cells. When coupling was insufficient to induce pacing, passive spread of the oscillations occurred in the follower. These results show a non-monotonic relationship between gK1, gf, gj, and pacing. Further, oscillations can be generated by gK1 and gf in the absence of SCN5A.


Subject(s)
Biological Clocks , Gap Junctions/physiology , Giant Cells/physiology , Membrane Potentials , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Giant Cells/cytology , HEK293 Cells , Humans
12.
Am J Physiol Heart Circ Physiol ; 319(5): H1112-H1122, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32986966

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) enable cardiotoxicity testing and personalized medicine. However, their maturity is of concern, including relatively depolarized resting membrane potential and more spontaneous activity compared with adult cardiomyocytes, implicating low or lacking inward rectifier potassium current (Ik1). Here, protein quantification confirms Kir2.1 expression in hiPSC-CM syncytia, albeit several times lower than in adult heart tissue. We find that hiPSC-CM culture density influences Kir2.1 expression at the mRNA level (potassium inwardly rectifying channel subfamily J member 2) and at the protein level and its associated electrophysiology phenotype. Namely, all-optical cardiac electrophysiology and pharmacological treatments reveal reduction of spontaneous and irregular activity and increase in action potential upstroke in denser cultures. Blocking Ik1-like currents with BaCl2 increased spontaneous frequency and blunted action potential upstrokes during pacing in a dose-dependent manner only in the highest-density cultures, in line with Ik1's role in regulating the resting membrane potential. Our results emphasize the importance of syncytial growth of hiPSC-CMs for more physiologically relevant phenotype and the power of all-optical electrophysiology to study cardiomyocytes in their multicellular setting.NEW & NOTEWORTHY We identify cell culture density and cell-cell contact as an important factor in determining the expression of a key ion channel at the transcriptional and the protein levels, KCNJ2/Kir2.1, and its contribution to the electrophysiology of human induced pluripotent stem cell-derived cardiomyocytes. Our results indicate that studies on isolated cells, out of tissue context, may underestimate the cellular ion channel properties being characterized.


Subject(s)
Giant Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Action Potentials , Animals , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques/methods , Cellular Reprogramming Techniques/standards , Female , Giant Cells/cytology , Giant Cells/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Primary Cell Culture/methods , Primary Cell Culture/standards , Rats
13.
Development ; 144(20): 3731-3743, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28935707

ABSTRACT

The vertebrate-specific ESCC microRNA family arises from two genetic loci in mammals: miR-290/miR-371 and miR-302. The miR-302 locus is found broadly among vertebrates, whereas the miR-290/miR-371 locus is unique to eutheria, suggesting a role in placental development. Here, we evaluate that role. A knock-in reporter for the mouse miR-290 cluster is expressed throughout the embryo until gastrulation, when it becomes specifically expressed in extraembryonic tissues and the germline. In the placenta, expression is limited to the trophoblast lineage, where it remains highly expressed until birth. Deletion of the miR-290 cluster gene (Mirc5) results in reduced trophoblast progenitor cell proliferation and a reduced DNA content in endoreduplicating trophoblast giant cells. The resulting placenta is reduced in size. In addition, the vascular labyrinth is disorganized, with thickening of the maternal-fetal blood barrier and an associated reduction in diffusion. Multiple mRNA targets of the miR-290 cluster microRNAs are upregulated. These data uncover a crucial function for the miR-290 cluster in the regulation of a network of genes required for placental development, suggesting a central role for these microRNAs in the evolution of placental mammals.


Subject(s)
MicroRNAs/genetics , MicroRNAs/physiology , Placenta/physiology , Animals , Cell Lineage , Cell Proliferation , Female , Gene Expression Profiling , Genotype , Giant Cells/cytology , Maternal-Fetal Exchange , Mice , Mice, Knockout , Multigene Family , Pregnancy , Sequence Analysis, RNA , Trophoblasts/metabolism
14.
New Phytol ; 225(1): 430-447, 2020 01.
Article in English | MEDLINE | ID: mdl-31505035

ABSTRACT

Galls induced by plant-parasitic nematodes involve a hyperactivation of the plant mitotic and endocycle machinery for their profit. Dedifferentiation of host root cells includes drastic cellular and molecular readjustments. In such a background, potential DNA damage in the genome of gall cells is evident. We investigated whether DNA damage checkpoint activation followed by DNA repair occurred, or was eventually circumvented, in nematode-induced galls. Galls display transcriptional activation of the DNA damage checkpoint kinase WEE1, correlated with its protein localization in the nuclei. The promoter of the stress marker gene SMR7 was evaluated under the WEE1-knockout background. Drugs inducing DNA damage and a marker for DNA repair, PARP1, were used to understand the mechanisms for coping with DNA damage in galls. Our functional study revealed that gall cells lacking WEE1 conceivably entered mitosis prematurely, disturbing the cell cycle despite the loss of genome integrity. The disrupted nuclei phenotype in giant cells hinted at the accumulation of mitotic defects. In addition, WEE1-knockout in Arabidopsis and downregulation in tomato repressed infection and reproduction of root-knot nematodes. Together with data on DNA-damaging drugs, we suggest a conserved function for WEE1 in controlling G1/S cell cycle arrest in response to a replication defect in galls.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Arabidopsis/parasitology , Cell Cycle , Plant Tumors/parasitology , Protein Serine-Threonine Kinases/metabolism , Tylenchoidea/physiology , Animals , Arabidopsis/genetics , Cell Cycle/genetics , Cell Nucleus/metabolism , DNA Damage , Gene Expression Regulation, Plant , Gene Knockout Techniques , Giant Cells/cytology , Glucuronidase/metabolism , Solanum lycopersicum/genetics , Mitosis , Plants, Genetically Modified , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Genes Dev ; 26(18): 2063-74, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22987638

ABSTRACT

Developmental arrest of Blimp1/Prdm1 mutant embryos at around embryonic day 10.5 (E10.5) has been attributed to placental disturbances. Here we investigate Blimp1/Prdm1 requirements in the trophoblast cell lineage. Loss of function disrupts specification of the invasive spiral artery-associated trophoblast giant cells (SpA-TGCs) surrounding maternal blood vessels and severely compromises the ability of the spongiotrophoblast layer to expand appropriately, secondarily causing collapse of the underlying labyrinth layer. Additionally, we identify a population of proliferating Blimp1(+) diploid cells present within the spongiotrophoblast layer. Lineage tracing experiments exploiting a novel Prdm1.Cre-LacZ allele demonstrate that these Blimp1(+) cells give rise to the mature SpA-TGCs, canal TGCs, and glycogen trophoblasts. In sum, the transcriptional repressor Blimp1/Prdm1 is required for terminal differentiation of SpA-TGCs and defines a lineage-restricted progenitor cell population contributing to placental growth and morphogenesis.


Subject(s)
Cell Differentiation , Giant Cells/cytology , Stem Cells/cytology , Transcription Factors/metabolism , Trophoblasts/cytology , Animals , Female , Gene Expression Profiling , Gene Expression Regulation , Mice , Placenta/blood supply , Placenta/cytology , Positive Regulatory Domain I-Binding Factor 1 , Pregnancy , Stem Cells/metabolism , Transcription Factors/genetics
16.
Int J Mol Sci ; 21(20)2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33086479

ABSTRACT

Classically, osteoclast fusion consists of four basic steps: (1) attraction/migration, (2) recognition, (3) cell-cell adhesion, and (4) membrane fusion. In theory, this sounds like a straightforward simple linear process. However, it is not. Osteoclast fusion has to take place in a well-coordinated manner-something that is not simple. In vivo, the complex regulation of osteoclast formation takes place within the bone marrow-in time and space. The present review will focus on considering osteoclast fusion in the context of physiology and pathology. Special attention is given to: (1) regulation of osteoclast fusion in vivo, (2) heterogeneity of osteoclast fusion partners, (3) regulation of multi-nucleation, (4) implications for physiology and pathology, and (5) implications for drug sensitivity and side effects. The review will emphasize that more attention should be given to the human in vivo reality when interpreting the impact of in vitro and animal studies. This should be done in order to improve our understanding of human physiology and pathology, as well as to improve anti-resorptive treatment and reduce side effects.


Subject(s)
Giant Cells/cytology , Osteoclasts/cytology , Animals , Cell Fusion , Humans , Membrane Fusion , Models, Animal , Proteins/metabolism
17.
Semin Cell Dev Biol ; 72: 45-55, 2017 12.
Article in English | MEDLINE | ID: mdl-29101004

ABSTRACT

The study of Drosophila muscle development dates back to the middle of the last century. Since that time, Drosophila has proved to be an ideal system for studying muscle development, differentiation, function, and disease. As in humans, Drosophila muscle forms via a series of conserved steps, starting with muscle specification, myoblast fusion, attachment to tendon cells, interactions with motorneurons, and sarcomere and myofibril formation. The genes and mechanisms required for these processes share striking similarities to those found in humans. The highly tractable genetic system and imaging approaches available in Drosophila allow for an efficient interrogation of muscle biology and for application of what we learn to other systems. In this article, we review our current understanding of muscle development in Drosophila, with a focus on myoblast fusion, the process responsible for the generation of syncytial muscle cells. We also compare and contrast those genes required for fusion in Drosophila and vertebrates.


Subject(s)
Cell Fusion , Giant Cells/cytology , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Animals , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Giant Cells/metabolism , Humans , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myoblasts/metabolism
18.
J Cell Sci ; 130(11): 1917-1928, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28424232

ABSTRACT

Cell-cell fusion is widely observed during development and disease, and imposes a dramatic change on participating cells. Cell fusion should be tightly controlled, but the underlying mechanism is poorly understood. Here, we found that the JAK/STAT pathway suppressed cell fusion during wound healing in the Drosophila larval epidermis, restricting cell fusion to the vicinity of the wound. In the absence of JAK/STAT signaling, a large syncytium containing a 3-fold higher number of nuclei than observed in wild-type tissue formed in wounded epidermis. The JAK/STAT ligand-encoding genes upd2 and upd3 were transcriptionally induced by wounding, and were required for suppressing excess cell fusion. JNK (also known as Basket in flies) was activated in the wound vicinity and activity peaked at ∼8 h after injury, whereas JAK/STAT signaling was activated in an adjoining concentric ring and activity peaked at a later stage. Cell fusion occurred primarily in the wound vicinity, where JAK/STAT activation was suppressed by fusion-inducing JNK signaling. JAK/STAT signaling was both necessary and sufficient for the induction of ßPS integrin (also known as Myospheroid) expression, suggesting that the suppression of cell fusion was mediated at least in part by integrin protein.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Epidermis/metabolism , Janus Kinases/genetics , Larva/metabolism , MAP Kinase Kinase 4/genetics , STAT Transcription Factors/genetics , Transcription Factors/genetics , Wound Healing/genetics , Animals , Cell Fusion , Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Epidermis/injuries , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Giant Cells/cytology , Giant Cells/metabolism , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Janus Kinases/metabolism , Larva/genetics , Larva/growth & development , MAP Kinase Kinase 4/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Transcription Factors/metabolism
19.
J Mater Sci Mater Med ; 30(6): 61, 2019 May 24.
Article in English | MEDLINE | ID: mdl-31127377

ABSTRACT

In vivo experiments are accompanied by ethical issues, including sacrificing a large number of animals as well as large costs. A new in vivo 3D screening system was developed to reduce the number of required animals without compromising the results. The present pilot study examined a multiwell array system in combination with three different collagen-based biomaterials (A, B and C) using subcutaneous implantation for 10 days and histological and histomorphometrical evaluations. The tissue reaction towards the device itself was dominated by mononuclear cells. However, three independent biomaterial-specific tissue reactions were observed in three chambers. The results showed a mononuclear cell-based tissue reaction in one chamber (A) and foreign body reaction by multinucleated giant cells in the other two chambers (B and C). Statistical analysis showed a significantly higher number of multinucleated giant cells in cases B and C than in case A (A vs. B; ***P < 0.001), (A vs. C; P < 0.01). These outcomes were comparable to previously published observations with conventional biomaterial implantation. The present data lead to the conclusion that this 3D screening system could be an alternative tool to enhance the effectiveness of in vivo experiments, thus offering a more economic strategy to screen biomaterial-related cellular reactions, while saving animals, without influencing the final outcome.


Subject(s)
Biocompatible Materials/chemistry , Cell Culture Techniques/instrumentation , Collagen/chemistry , Foreign-Body Reaction , Animals , Giant Cells/cytology , Immune System , Inflammation , Mice , Pilot Projects , Skin/metabolism , Swine , Tissue Engineering/methods , Translational Research, Biomedical
20.
Int J Mol Sci ; 20(18)2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31540219

ABSTRACT

During the peri-implantation period, multinucleated syncytia are formed in the sheep placenta. For over 20 years the scientific consensus has been that during trophoblast syncytialization in sheep, binucleate trophoblast giant cells (BNCs) differentiate from mononuclear trophoblast cells, and individual BNCs fuse with individual luminal epithelial (LE) cells to form trinucleate cells. These trophoblast-LE syncytial plaques then grow through continued BNC migration and fusion. Therefore, LE cells are thought to be incorporated into syncytial plaques. However, these ideas were based on electron microscopy studies, without benefit of molecular markers for BNC and LE cells to support conclusions. The aim of this study was to observe interactions between BNCs and uterine LE cells using immunohistochemical localization for molecular markers for BNCs and uterine LE cells. We performed immunofluorescence staining, laser capture microdissection, and TUNEL staining on the uterine-placental tissues of sheep during early placentation. We observed: (1) syncytial cells containing more than two nuclei within the trophoblast cell layer; (2) depolarized LE cells that express caspase 3 and stain positively for TUNEL; (3) engulfment of caspase 3-positive LE cells by trophoblast giant cells (TGCs) and empty spaces within the LE layer at sites of implantation; (4) rapid enlargement of syncytial plaques; and (5) E-cadherin and TUNEL-positive cells within the uterine stroma underlying degenerating LE was coincident with accumulation of CD45-positive cells at these sites. These data suggest that during early placentation: (1) fusion between trophoblasts is not limited to the formation of BNCs, and the term 'trophoblast giant cell (TGC)' may be appropriate; (2) LE cells undergo apoptosis; (3) apoptotic LE cells are eliminated by TGCs; (4) fusion is not limited to the incorporation of new BNCs but involves the lateral fusion between growing syncytial plaques; and (5) TGCs carry apoptotic LE cells away from the uterine-placental interface for elimination by immune cells within the stroma. These data indicate that uterine LE cells are not incorporated into syncytial plaques, but are engulfed and eliminated, and that early placentation in sheep is more similar to early placentation in humans than is currently understood in that both develop mononucleated cytotrophoblast and multinucleated syncytiotrophoblast layers of entirely placental origin. The elimination of LE cells by sheep TGCs might provide insights into elimination and penetration of LE cells during human embryo implantation.


Subject(s)
Biomarkers/metabolism , Epithelial Cells/cytology , Giant Cells/cytology , Placentation , Trophoblasts/cytology , Animals , Cadherins/metabolism , Caspase 3/metabolism , Cell Differentiation , Cell Fusion , Cell Movement , Epithelial Cells/metabolism , Female , Giant Cells/metabolism , Immunohistochemistry , Keratins/metabolism , Leukocyte Common Antigens/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy , Sheep , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL