Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 757
Filter
1.
Cells ; 12(9)2023 05 07.
Article in English | MEDLINE | ID: mdl-37174735

ABSTRACT

Centrosome formation during early development in mice and rats occurs due to the appearance of centrioles de novo. In contrast, in humans and other non-rodent mammals, centrioles are thought to be derived from spermatozoa. Ultrastructural study of zygotes and early embryos of cattle at full series of ultrathin sections show that the proximal centriole of the spermatozoon disappears by the end of the first cleavage division. Centrioles appear in two to four cell embryos in fertilized oocytes and in parthenogenetic embryos. Centriole formation includes the appearance of atypical centrioles with randomly arranged triplets and centrioles with microtubule triplets of various lengths. After the third cleavage, four centriolar cylinders appear for the first time in the blastomeres while each embryo still has two atypical centrioles. Our results showed that the mechanisms of centriole formation in different groups of mammals are universal, differing only in the stage of development in which they occur.


Subject(s)
Centrosome , Oocytes , Humans , Male , Cattle , Animals , Mice , Rats , Oocytes/ultrastructure , Centrosome/ultrastructure , Centrioles/ultrastructure , Spermatozoa/ultrastructure , Mammals
2.
Adv Anat Embryol Cell Biol ; 235: 37-42, 2022.
Article in English | MEDLINE | ID: mdl-36525109

ABSTRACT

Among the multiple and intriguing roles of centrosomes in cellular functions is the ubiquitin-proteasome-mediated protein degradation. It has been shown that proteasomes are concentrated at the mammalian centrosome which led to further studies to view the centrosome as a proteolytic center (Wojcik et al. 1996; Wigley et al. 1999; reviewed in Badano et al. 2005). Proteasomal components that are concentrated around the centrosome include ubiquitin, the 20S and 19S subunits of the proteasome, as well as the E3 enzyme parkin. These proteasomal components colocalize with the centrosomal marker γ-tubulin and co-purify with γ-tubulin in the centrosomal fractions after sucrose-gradient ultracentrifugation (Wigley et al. 1999). The localization, accumulation, and concentration of proteasomal components around centrosomes appear to be microtubule independent which has been shown experimentally by inhibiting microtubule functions. When intracellular levels of misfolded proteins were experimentally increased by either proteasome inhibition with drugs such as lactacystin, or by overexpression of misfolded mutant proteins, the centrosome-associated proteasome network became expanded and proteolytic components were recruited from the cytosol without involvement of microtubules. These studies revealed a critical role of centrosomes in the organization and subcellular localization of proteasomes (Wigley et al. 1999; Fabunmi et al. 2000).


Subject(s)
Proteasome Endopeptidase Complex , Tubulin , Animals , Humans , Tubulin/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Centrosome/metabolism , Centrosome/ultrastructure , Ubiquitin/metabolism , Mammals/metabolism
3.
Cell Rep Methods ; 2(11): 100322, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36452870

ABSTRACT

Electron microscopy is the gold standard to characterize centrosomal ultrastructure. However, production of significant morphometrical data is highly limited by acquisition time. We therefore developed a generalizable, semi-automated high-throughput electron tomography strategy to study centrosome aberrations in sparse patient-derived cancer cells at nanoscale. As proof of principle, we present electron tomography data on 455 centrioles of CD138pos plasma cells from one patient with relapsed/refractory multiple myeloma and CD138neg bone marrow mononuclear cells from three healthy donors as a control. Plasma cells from the myeloma patient displayed 122 over-elongated centrioles (48.8%). Particularly mother centrioles also harbored gross structural abnormalities, including fragmentation and disturbed microtubule cylinder formation, while control centrioles were phenotypically unremarkable. These data demonstrate the feasibility of our scalable high-throughput electron tomography strategy to study structural centrosome aberrations in primary tumor cells. Moreover, our electron tomography workflow and data provide a resource for the characterization of cell organelles beyond centrosomes.


Subject(s)
Centrioles , Multiple Myeloma , Humans , Centrioles/pathology , Multiple Myeloma/diagnostic imaging , Electron Microscope Tomography , Workflow , Centrosome/ultrastructure
4.
Cells ; 11(5)2022 02 22.
Article in English | MEDLINE | ID: mdl-35269380

ABSTRACT

The sperm competition theory, as proposed by Geoff Parker, predicts that sperm evolve through a cascade of changes. As an example, internal fertilization is followed by sperm morphology diversification. However, little is known about the evolution of internal sperm structures. The centriole has an ancient and evolutionarily conserved canonical structure with signature 9-fold, radially symmetric microtubules that form the cell's centrosomes, cilia, and flagella. Most animal spermatozoa have two centrioles, one of which forms the spermatozoan flagellum. Both are delivered to the egg and constitute the embryo's first two centrosomes. The spermatozoa of mammals and insects only have one recognizable centriole with a canonical structure. A second sperm centriole with an atypical structure was recently reported in both animal groups and which, prior to this, eluded discovery by standard techniques and criteria. Because the ancestors of both mammals and insects reproduced by internal fertilization, we hypothesized that the transition from two centrioles with canonical composition in ancestral sperm to an atypical centriolar composition characterized by only one canonical centriole evolved preferentially after internal fertilization. We examined fish because of the diversity of species available to test this hypothesis−as some species reproduce via internal and others via external fertilization−and because their spermatozoan ultrastructure has been extensively studied. Our literature search reports on 277 fish species. Species reported with atypical centriolar composition are specifically enriched among internal fertilizers compared to external fertilizers (7/34, 20.6% versus 2/243, 0.80%; p < 0.00001, odds ratio = 32.4) and represent phylogenetically unrelated fish. Atypical centrioles are present in the internal fertilizers of the subfamily Poeciliinae. Therefore, internally fertilizing fish preferentially and independently evolved spermatozoa with atypical centriolar composition multiple times, agreeing with Parker's cascade theory.


Subject(s)
Centrioles , Fertilizers , Animals , Centrioles/ultrastructure , Centrosome/ultrastructure , Fertilization , Male , Mammals , Spermatozoa/ultrastructure
5.
Nat Commun ; 12(1): 6042, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654813

ABSTRACT

Centriole biogenesis and maintenance are crucial for cells to generate cilia and assemble centrosomes that function as microtubule organizing centers (MTOCs). Centriole biogenesis and MTOC function both require the microtubule nucleator γ-tubulin ring complex (γTuRC). It is widely accepted that γTuRC nucleates microtubules from the pericentriolar material that is associated with the proximal part of centrioles. However, γTuRC also localizes more distally and in the centriole lumen, but the significance of these findings is unclear. Here we identify spatially and functionally distinct subpopulations of centrosomal γTuRC. Luminal localization is mediated by augmin, which is linked to the centriole inner scaffold through POC5. Disruption of luminal localization impairs centriole integrity and interferes with cilium assembly. Defective ciliogenesis is also observed in γTuRC mutant fibroblasts from a patient suffering from microcephaly with chorioretinopathy. These results identify a non-canonical role of augmin-γTuRC in the centriole lumen that is linked to human disease.


Subject(s)
Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/isolation & purification , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins/ultrastructure , Cell Line , Centrioles/ultrastructure , Centrosome/metabolism , Centrosome/ultrastructure , Cilia , Female , Humans , Male , Mice , Microtubule-Associated Proteins/ultrastructure , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Neurons
6.
Cells ; 10(10)2021 10 05.
Article in English | MEDLINE | ID: mdl-34685637

ABSTRACT

The centrosome of Dictyostelium amoebae contains no centrioles and consists of a cylindrical layered core structure surrounded by a corona harboring microtubule-nucleating γ-tubulin complexes. It is the major centrosomal model beyond animals and yeasts. Proteomics, protein interaction studies by BioID and superresolution microscopy methods led to considerable progress in our understanding of the composition, structure and function of this centrosome type. We discuss all currently known components of the Dictyostelium centrosome in comparison to other centrosomes of animals and yeasts.


Subject(s)
Centrosome/metabolism , Dictyostelium/metabolism , Cell Nucleus/metabolism , Centrosome/ultrastructure , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
7.
Cells ; 10(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34440693

ABSTRACT

With the advent of super-resolution microscopy, we gained a powerful toolbox to bridge the gap between the cellular- and molecular-level analysis of living organisms. Although nanoscopy is broadly applicable, classical model organisms, such as fruit flies, worms and mice, remained the leading subjects because combining the strength of sophisticated genetics, biochemistry and electrophysiology with the unparalleled resolution provided by super-resolution imaging appears as one of the most efficient approaches to understanding the basic cell biological questions and the molecular complexity of life. Here, we summarize the major nanoscopic techniques and illustrate how these approaches were used in Drosophila model systems to revisit a series of well-known cell biological phenomena. These investigations clearly demonstrate that instead of simply achieving an improvement in image quality, nanoscopy goes far beyond with its immense potential to discover novel structural and mechanistic aspects. With the examples of synaptic active zones, centrosomes and sarcomeres, we will explain the instrumental role of super-resolution imaging pioneered in Drosophila in understanding fundamental subcellular constituents.


Subject(s)
Drosophila/ultrastructure , Microscopy, Fluorescence/methods , Models, Biological , Single Molecule Imaging/methods , Animals , Centrosome/metabolism , Centrosome/ultrastructure , Drosophila/metabolism , Sarcomeres/metabolism , Sarcomeres/ultrastructure
8.
Nat Commun ; 12(1): 4096, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215746

ABSTRACT

Non-centrosomal microtubule arrays serve crucial functions in cells, yet the mechanisms of their generation are poorly understood. During budding of the epithelial tubes of the salivary glands in the Drosophila embryo, we previously demonstrated that the activity of pulsatile apical-medial actomyosin depends on a longitudinal non-centrosomal microtubule array. Here we uncover that the exit from the last embryonic division cycle of the epidermal cells of the salivary gland placode leads to one centrosome in the cells losing all microtubule-nucleation capacity. This restriction of nucleation activity to the second, Centrobin-enriched, centrosome is key for proper morphogenesis. Furthermore, the microtubule-severing protein Katanin and the minus-end-binding protein Patronin accumulate in an apical-medial position only in placodal cells. Loss of either in the placode prevents formation of the longitudinal microtubule array and leads to loss of apical-medial actomyosin and impaired apical constriction. We thus propose a mechanism whereby Katanin-severing at the single active centrosome releases microtubule minus-ends that are then anchored by apical-medial Patronin to promote formation of the longitudinal microtubule array crucial for apical constriction and tube formation.


Subject(s)
Cell Division/physiology , Centrosome/metabolism , Microtubules/metabolism , Actins , Actomyosin/metabolism , Animals , Centrosome/ultrastructure , Cytoskeletal Proteins/metabolism , Drosophila , Katanin , Male , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , Morphogenesis , Salivary Glands , Tubulin/metabolism
9.
Cancer Sci ; 112(5): 1679-1687, 2021 May.
Article in English | MEDLINE | ID: mdl-33606355

ABSTRACT

Alterations in breast cancer gene 1 (BRCA1), a tumor suppressor gene, increase the risk of breast and ovarian cancers. BRCA1 forms a heterodimer with BRCA1-associated RING domain protein 1 (BARD1) and functions in multiple cellular processes, including DNA repair and centrosome regulation. BRCA1 acts as a tumor suppressor by promoting homologous recombination (HR) repair, and alterations in BRCA1 cause HR deficiency, not only in breast and ovarian tissues but also in other tissues. The molecular mechanisms underlying BRCA1 alteration-induced carcinogenesis remain unclear. Centrosomes are the major microtubule-organizing centers and function in bipolar spindle formation. The regulation of centrosome number is critical for chromosome segregation in mitosis, which maintains genomic stability. BRCA1/BARD1 function in centrosome regulation together with Obg-like ATPase (OLA1) and receptor for activating protein C kinase 1 (RACK1). Cancer-derived variants of BRCA1, BARD1, OLA1, and RACK1 do not interact, and aberrant expression of these proteins results in abnormal centrosome duplication in mammary-derived cells, and rarely in other cell types. RACK1 is involved in centriole duplication in the S phase by promoting polo-like kinase 1 activation by Aurora A, which is critical for centrosome duplication. Centriole number is higher in cells derived from mammary tissues compared with in those derived from other tissues, suggesting that tissue-specific centrosome characterization may shed light on the tissue specificity of BRCA1-associated carcinogenesis. Here, we explored the role of the BRCA1-containing complex in centrosome regulation and the effect of its deficiency on tissue-specific carcinogenesis.


Subject(s)
BRCA1 Protein/deficiency , Carcinogenesis/metabolism , Centrosome/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases/metabolism , BRCA1 Protein/chemistry , BRCA1 Protein/metabolism , Carcinogenesis/genetics , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Centrosome/ultrastructure , Chromosomal Instability , Female , GTP-Binding Proteins/metabolism , Genes, BRCA1 , Humans , Mitosis/genetics , Neoplasm Proteins/metabolism , Organ Specificity , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors for Activated C Kinase/metabolism , Recombinational DNA Repair , Spindle Apparatus/genetics , Tumor Suppressor Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Polo-Like Kinase 1
10.
J Cell Biol ; 220(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33475699

ABSTRACT

Primary cilia protrude from the cell surface and have diverse roles during development and disease, which depends on the precise timing and control of cilia assembly and disassembly. Inactivation of assembly often causes cilia defects and underlies ciliopathy, while diseases caused by dysfunction in disassembly remain largely unknown. Here, we demonstrate that CEP55 functions as a cilia disassembly regulator to participate in ciliopathy. Cep55-/- mice display clinical manifestations of Meckel-Gruber syndrome, including perinatal death, polycystic kidneys, and abnormalities in the CNS. Interestingly, Cep55-/- mice exhibit an abnormal elongation of cilia on these tissues. Mechanistically, CEP55 promotes cilia disassembly by interacting with and stabilizing Aurora A kinase, which is achieved through facilitating the chaperonin CCT complex to Aurora A. In addition, CEP55 mutation in Meckel-Gruber syndrome causes the failure of cilia disassembly. Thus, our study establishes a cilia disassembly role for CEP55 in vivo, coupling defects in cilia disassembly to ciliopathy and further suggesting that proper cilia dynamics are critical for mammalian development.


Subject(s)
Aurora Kinase A/metabolism , Cell Cycle Proteins/metabolism , Cilia/metabolism , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/deficiency , Cells, Cultured , Centrosome/metabolism , Centrosome/ultrastructure , Chaperonin Containing TCP-1/metabolism , Cilia/ultrastructure , Ciliary Motility Disorders/pathology , Encephalocele/pathology , Enzyme Stability , Gene Targeting , HEK293 Cells , Humans , Mice , Mitosis , Phenotype , Polycystic Kidney Diseases/pathology , Protein Binding , Retinitis Pigmentosa/pathology , Smoothened Receptor/metabolism
11.
Semin Cell Dev Biol ; 110: 89-103, 2021 02.
Article in English | MEDLINE | ID: mdl-32540122

ABSTRACT

Primary cilia are immotile appendages that have evolved to receive and interpret a variety of different extracellular cues. Cilia play crucial roles in intercellular communication during development and defects in cilia affect multiple tissues accounting for a heterogeneous group of human diseases called ciliopathies. The Hedgehog (Hh) signaling pathway is one of these cues and displays a unique and symbiotic relationship with cilia. Not only does Hh signaling require cilia for its function but the majority of the Hh signaling machinery is physically located within the cilium-centrosome complex. More specifically, cilia are required for both repressing and activating Hh signaling by modifying bifunctional Gli transcription factors into repressors or activators. Defects in balancing, interpreting or establishing these repressor/activator gradients in Hh signaling either require cilia or phenocopy disruption of cilia. Here, we will summarize the current knowledge on how spatiotemporal control of the molecular machinery of the cilium allows for a tight control of basal repression and activation states of the Hh pathway. We will then discuss several paradigms on how cilia influence Hh pathway activity in tissue morphogenesis during development. Last, we will touch on how cilia and Hh signaling are being reactivated and repurposed during adult tissue regeneration. More specifically, we will focus on mesenchymal stem cells within the connective tissue and discuss the similarities and differences of how cilia and ciliary Hh signaling control the formation of fibrotic scar and adipose tissue during fatty fibrosis of several tissues.


Subject(s)
Cilia/metabolism , Ciliopathies/genetics , Hedgehog Proteins/genetics , Obesity/genetics , Regeneration/genetics , Zinc Finger Protein GLI1/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Centrosome/metabolism , Centrosome/ultrastructure , Cilia/pathology , Cilia/ultrastructure , Ciliopathies/metabolism , Ciliopathies/pathology , Connective Tissue/metabolism , Connective Tissue/pathology , Fibrosis , Gene Expression Regulation , Hedgehog Proteins/metabolism , Humans , Light Signal Transduction , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Microtubules/metabolism , Microtubules/ultrastructure , Morphogenesis/genetics , Obesity/metabolism , Obesity/pathology , Zinc Finger Protein GLI1/metabolism
12.
Semin Cell Dev Biol ; 110: 113-122, 2021 02.
Article in English | MEDLINE | ID: mdl-32409142

ABSTRACT

Centrosomes are the major microtubule organizing centers in a large number of animal cells. They are involved in diverse cellular functions like cell division, migration, sensing and motility. Despite being identified more than 100 years ago, they did not receive much attention until recent discoveries suggesting their association with human diseases. Centrosome-related defects have been observed in several human diseases including cancers, brain disorders and ciliopathies. Researchers in the field are trying to understand the relationship between centrosomes and these diseases. Accordingly, this review provides an overview of the current knowledge regarding the role of centrosomes during ciliogenesis and neural stem cell division. The review primarily focuses on the impairment of centrosome number, organization and functioning leading to a wide range of human diseases. Finally, we discuss the scope of targeting centrosomes for therapeutic purposes.


Subject(s)
Centrosome/metabolism , Cilia/metabolism , Ciliopathies/genetics , Neoplasms/genetics , Nerve Tissue Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Centrosome/drug effects , Centrosome/pathology , Centrosome/ultrastructure , Cilia/drug effects , Cilia/pathology , Cilia/ultrastructure , Ciliopathies/metabolism , Ciliopathies/pathology , Gene Expression Regulation , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Microtubules/metabolism , Microtubules/ultrastructure , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phthalazines/pharmacology , Pyrimidines/pharmacology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Signal Transduction
13.
Semin Cell Dev Biol ; 110: 61-69, 2021 02.
Article in English | MEDLINE | ID: mdl-32307225

ABSTRACT

Cilia and centrosomes of eukaryotic cells play important roles in cell movement, fluid transport, extracellular sensing, and chromosome division. The physiological functions of cilia and centrosomes are generated by their dynamics, motions, and forces controlled by the physical, chemical, and biological environments. How an individual cilium achieves its beat pattern and induces fluid flow is governed by its ultrastructure as well as the coordination of associated molecular motors. Thus, a bottom-up understanding of the physiological functions of cilia and centrosomes from the molecular to tissue levels is required. Correlations between the structure and motion can be understood in terms of mechanics. This review first focuses on cilia and centrosomes at the molecular level, introducing their ultrastructure. We then shift to the organelle level and introduce the kinematics and mechanics of cilia and centrosomes. Next, at the tissue level, we introduce nodal ciliary dynamics and nodal flow, which play crucial roles in the organogenetic process of left-right asymmetry. We also introduce respiratory ciliary dynamics and mucous flow, which are critical for protecting the epithelium from drying and exposure to harmful particles and viruses, i.e., respiratory clearance function. Finally, we discuss the future research directions in this field.


Subject(s)
Axoneme/ultrastructure , Basal Bodies/ultrastructure , Centrosome/ultrastructure , Cilia/ultrastructure , Epithelial Cells/ultrastructure , Microtubules/ultrastructure , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism , Axoneme/metabolism , Basal Bodies/metabolism , Biological Transport , Biomechanical Phenomena , Centrosome/metabolism , Chromosome Segregation , Cilia/metabolism , Epithelial Cells/metabolism , Gene Expression , Humans , Microtubules/metabolism , Movement , Organogenesis/genetics , Respiration/genetics , Rheology
15.
Cells ; 9(10)2020 09 24.
Article in English | MEDLINE | ID: mdl-32987651

ABSTRACT

The centrosome, which consists of two centrioles surrounded by pericentriolar material, is a unique structure that has retained its main features in organisms of various taxonomic groups from unicellular algae to mammals over one billion years of evolution. In addition to the most noticeable function of organizing the microtubule system in mitosis and interphase, the centrosome performs many other cell functions. In particular, centrioles are the basis for the formation of sensitive primary cilia and motile cilia and flagella. Another principal function of centrosomes is the concentration in one place of regulatory proteins responsible for the cell's progression along the cell cycle. Despite the existing exceptions, the functioning of the centrosome is subject to general principles, which are discussed in this review.


Subject(s)
Centrosome/metabolism , Actin Cytoskeleton/metabolism , Animals , Basal Bodies/metabolism , Centrioles/metabolism , Centrioles/ultrastructure , Centrosome/ultrastructure , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Models, Biological
16.
Biochem Soc Trans ; 48(5): 2101-2115, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32897294

ABSTRACT

Microcephaly is a rare, yet devastating, neurodevelopmental condition caused by genetic or environmental insults, such as the Zika virus infection. Microcephaly manifests with a severely reduced head circumference. Among the known heritable microcephaly genes, a significant proportion are annotated with centrosome-related ontologies. Centrosomes are microtubule-organizing centers, and they play fundamental roles in the proliferation of the neuronal progenitors, the neural stem cells (NSCs), which undergo repeated rounds of asymmetric cell division to drive neurogenesis and brain development. Many of the genes, pathways, and developmental paradigms that dictate NSC development in humans are conserved in Drosophila melanogaster. As such, studies of Drosophila NSCs lend invaluable insights into centrosome function within NSCs and help inform the pathophysiology of human microcephaly. This mini-review will briefly survey causative links between deregulated centrosome functions and microcephaly with particular emphasis on insights learned from Drosophila NSCs.


Subject(s)
Centrosome/ultrastructure , Drosophila melanogaster/metabolism , Neural Stem Cells/metabolism , Animals , Brain/embryology , Brain/growth & development , Cell Division , Cell Proliferation , Disease Models, Animal , Humans , Microcephaly/metabolism , Microtubule-Organizing Center/metabolism , Mitosis , Neurogenesis , Neurons , Stem Cells/cytology , Zika Virus , Zika Virus Infection/metabolism
17.
Biochem Soc Trans ; 48(5): 1929-1939, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32897366

ABSTRACT

The OFD1 gene was initially identified as the gene responsible for the X-linked dominant male lethal OFD type I syndrome, a developmental disorder ascribed to cilia disfunction. The transcript has been subsequently associated to four different X-linked recessive conditions, namely Joubert syndrome, retinitis pigmentosa, primary ciliary dyskinesia and Simpson-Golabi-Behmel type 2 syndrome. The centrosomal/basal body OFD1 protein has indeed been shown to be required for primary cilia formation and left-right asymmetry. The protein is also involved in other tasks, e.g. regulation of cellular protein content, constrain of the centriolar length, chromatin remodeling at DNA double strand breaks, control of protein quality balance and cell cycle progression, which might be mediated by non-ciliary activities. OFD1 represents a paradigmatic model of a protein that performs its diverse actions according to the cell needs and depending on the subcellular localization, the cell type/tissue and other possible factors still to be determined. An increased number of multitask protein, such as OFD1, may represent a partial explanation to human complexity, as compared with less complex organisms with an equal or slightly lower number of proteins.


Subject(s)
Cell Cycle , Chromosomes, Human, X , Cilia/metabolism , DNA Breaks, Double-Stranded , Mutation , Proteins/genetics , Abnormalities, Multiple/genetics , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Centrosome/ultrastructure , Cerebellum/abnormalities , Chromatin/metabolism , Ciliary Motility Disorders/genetics , Cytoplasm/metabolism , Eye Abnormalities/genetics , Genetic Diseases, X-Linked/genetics , Humans , Kidney/metabolism , Kidney Diseases, Cystic/genetics , Phenotype , Rare Diseases/genetics , Retina/abnormalities , Retinitis Pigmentosa/genetics
18.
Biochimie ; 177: 127-131, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32841682

ABSTRACT

A-kinase anchoring protein 350 (AKAP350) is a centrosomal/Golgi scaffold protein, critical for the regulation of microtubule dynamics. AKAP350 recruits end-binding protein 1 (EB1) to the centrosome in mitotic cells, ensuring proper spindle orientation in epithelial cells. AKAP350 also interacts with p150glued, the main component of the dynactin complex. In the present work, we found that AKAP350 localized p150glued to the spindle poles, facilitating p150glued/EB1 interaction at these structures. Our results further showed that the decrease in AKAP350 expression reduced p150glued localization at astral microtubules and impaired the elongation of astral microtubules during anaphase. Overall, this study provides mechanistic data on how microtubule regulatory proteins gather to define microtubule dynamics in mitotic cells.


Subject(s)
A Kinase Anchor Proteins/physiology , Dynactin Complex/physiology , Spindle Poles/metabolism , Animals , Centrosome/metabolism , Centrosome/ultrastructure , Dogs , Madin Darby Canine Kidney Cells , Microtubules/metabolism , Microtubules/ultrastructure , Spindle Poles/ultrastructure
19.
PLoS Biol ; 18(8): e3000762, 2020 08.
Article in English | MEDLINE | ID: mdl-32760088

ABSTRACT

Centrosomes, the main microtubule organizing centers (MTOCs) of metazoan cells, contain an older "mother" and a younger "daughter" centriole. Stem cells either inherit the mother or daughter-centriole-containing centrosome, providing a possible mechanism for biased delivery of cell fate determinants. However, the mechanisms regulating centrosome asymmetry and biased centrosome segregation are unclear. Using 3D-structured illumination microscopy (3D-SIM) and live-cell imaging, we show in fly neural stem cells (neuroblasts) that the mitotic kinase Polo and its centriolar protein substrate Centrobin (Cnb) accumulate on the daughter centriole during mitosis, thereby generating molecularly distinct mother and daughter centrioles before interphase. Cnb's asymmetric localization, potentially involving a direct relocalization mechanism, is regulated by Polo-mediated phosphorylation, whereas Polo's daughter centriole enrichment requires both Wdr62 and Cnb. Based on optogenetic protein mislocalization experiments, we propose that the establishment of centriole asymmetry in mitosis primes biased interphase MTOC activity, necessary for correct spindle orientation.


Subject(s)
Cell Cycle Proteins/genetics , Centrioles/metabolism , Centrosome/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Mitosis , Protein Serine-Threonine Kinases/genetics , Animals , Animals, Genetically Modified , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Cycle Proteins/metabolism , Centrioles/ultrastructure , Centrosome/ultrastructure , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interphase , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Optogenetics/methods , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Red Fluorescent Protein
20.
FEBS J ; 287(17): 3672-3676, 2020 09.
Article in English | MEDLINE | ID: mdl-32692465

ABSTRACT

The novel coronavirus SARS-CoV-2 is the causative agent of the global coronavirus disease 2019 (COVID-19) outbreak. In addition to pneumonia, other COVID-19-associated symptoms have been reported, including loss of smell (anosmia). However, the connection between infection with coronavirus and anosmia remains enigmatic. It has been reported that defects in olfactory cilia lead to anosmia. In this Viewpoint, we summarize transmission electron microscopic studies of cilia in virus-infected cells. In the human nasal epithelium, coronavirus infects the ciliated cells and causes deciliation. Research has shown that viruses such as influenza and Sendai attach to the ciliary membrane. The Sendai virus enters cilia by fusing its viral membrane with the ciliary membrane. A recent study on SARS-CoV-2-human protein-protein interactions revealed that the viral nonstructural protein Nsp13 interacts with the centrosome components, providing a potential molecular link. The mucociliary escalator removes inhaled pathogenic particles and functions as the first line of protection mechanism against viral infection in the human airway. Thus, future investigation into the virus-cilium interface will help further the battle against COVID-19.


Subject(s)
Anosmia/metabolism , COVID-19/metabolism , Centrosome/virology , Cilia/virology , Nasal Mucosa/virology , SARS-CoV-2/pathogenicity , Viral Nonstructural Proteins/metabolism , Anosmia/complications , Anosmia/physiopathology , Anosmia/virology , COVID-19/complications , COVID-19/physiopathology , COVID-19/virology , Centrosome/metabolism , Centrosome/ultrastructure , Cilia/metabolism , Cilia/ultrastructure , Host-Pathogen Interactions/genetics , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nasal Mucosa/metabolism , Nasal Mucosa/ultrastructure , Orthomyxoviridae/metabolism , Orthomyxoviridae/pathogenicity , Protein Binding , RNA Helicases/genetics , RNA Helicases/metabolism , SARS-CoV-2/metabolism , Sendai virus/metabolism , Sendai virus/pathogenicity , Severity of Illness Index , Smell/physiology , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...