Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
EMBO J ; 40(17): e108498, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34309047

RESUMEN

Tubulin polyglutamylation is a post-translational modification of the microtubule cytoskeleton, which is generated by a variety of enzymes with different specificities. The "tubulin code" hypothesis predicts that modifications generated by specific enzymes selectively control microtubule functions. Our recent finding that excessive accumulation of polyglutamylation in neurons causes their degeneration and perturbs axonal transport provides an opportunity for testing this hypothesis. By developing novel mouse models and a new glutamylation-specific antibody, we demonstrate here that the glutamylases TTLL1 and TTLL7 generate unique and distinct glutamylation patterns on neuronal microtubules. We find that under physiological conditions, TTLL1 polyglutamylates α-tubulin, while TTLL7 modifies ß-tubulin. TTLL1, but not TTLL7, catalyses the excessive hyperglutamylation found in mice lacking the deglutamylase CCP1. Consequently, deletion of TTLL1, but not of TTLL7, prevents degeneration of Purkinje cells and of myelinated axons in peripheral nerves in these mice. Moreover, loss of TTLL1 leads to increased mitochondria motility in neurons, while loss of TTLL7 has no such effect. By revealing how specific patterns of tubulin glutamylation, generated by distinct enzymes, translate into specific physiological and pathological readouts, we demonstrate the relevance of the tubulin code for homeostasis.


Asunto(s)
Transporte Axonal , Enfermedades Neurodegenerativas/metabolismo , Péptido Sintasas/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Péptido Sintasas/genética , Ácido Poliglutámico/metabolismo , Células de Purkinje/metabolismo
2.
EMBO J ; 37(23)2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30420556

RESUMEN

Posttranslational modifications of tubulin are emerging regulators of microtubule functions. We have shown earlier that upregulated polyglutamylation is linked to rapid degeneration of Purkinje cells in mice with a mutation in the deglutamylating enzyme CCP1. How polyglutamylation leads to degeneration, whether it affects multiple neuron types, or which physiological processes it regulates in healthy neurons has remained unknown. Here, we demonstrate that excessive polyglutamylation induces neurodegeneration in a cell-autonomous manner and can occur in many parts of the central nervous system. Degeneration of selected neurons in CCP1-deficient mice can be fully rescued by simultaneous knockout of the counteracting polyglutamylase TTLL1. Excessive polyglutamylation reduces the efficiency of neuronal transport in cultured hippocampal neurons, suggesting that impaired cargo transport plays an important role in the observed degenerative phenotypes. We thus establish polyglutamylation as a cell-autonomous mechanism for neurodegeneration that might be therapeutically accessible through manipulation of the enzymes that control this posttranslational modification.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional , Células de Purkinje/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Transporte Biológico Activo/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Péptidos/genética , Células de Purkinje/patología , Tubulina (Proteína)/genética
3.
J Cell Sci ; 133(3)2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31932508

RESUMEN

Neurons are highly complex cells that heavily rely on intracellular transport to distribute a range of functionally essential cargoes within the cell. Post-translational modifications of tubulin are emerging as mechanisms for regulating microtubule functions, but their impact on neuronal transport is only marginally understood. Here, we have systematically studied the impact of post-translational polyglutamylation on axonal transport. In cultured hippocampal neurons, deletion of a single deglutamylase, CCP1 (also known as AGTPBP1), is sufficient to induce abnormal accumulation of polyglutamylation, i.e. hyperglutamylation. We next investigated how hyperglutamylation affects axonal transport of a range of functionally different neuronal cargoes: mitochondria, lysosomes, LAMP1 endosomes and BDNF vesicles. Strikingly, we found a reduced motility for all these cargoes, suggesting that polyglutamylation could act as a regulator of cargo transport in neurons. This, together with the recent discovery that hyperglutamylation induces neurodegeneration, makes it likely that perturbed neuronal trafficking could be one of the central molecular causes underlying this novel type of degeneration.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Neuronas , Tubulina (Proteína) , Transporte Axonal , Hipocampo/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo
5.
J Cell Sci ; 132(3)2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30635446

RESUMEN

Sperm cells are highly specialized mammalian cells, and their biogenesis requires unique intracellular structures. Perturbation of spermatogenesis often leads to male infertility. Here, we assess the role of a post-translational modification of tubulin, glutamylation, in spermatogenesis. We show that mice lacking the tubulin deglutamylase CCP5 (also known as AGBL5) do not form functional sperm. In these mice, spermatids accumulate polyglutamylated tubulin, accompanied by the occurrence of disorganized microtubule arrays, in particular in the sperm manchette. Spermatids further fail to re-arrange their intracellular space and accumulate organelles and cytosol, while nuclei condense normally. Strikingly, spermatids lacking CCP5 show supernumerary centrioles, suggesting that glutamylation could control centriole duplication. We show that most of these observed defects are also present in mice in which CCP5 is deleted only in the male germ line, strongly suggesting that they are germ-cell autonomous. Our findings reveal that polyglutamylation is, beyond its known importance for sperm flagella, an essential regulator of several microtubule-based functions during spermatogenesis. This makes enzymes involved in glutamylation prime candidates for being genes involved in male sterility.


Asunto(s)
Carboxipeptidasas/genética , Infertilidad Masculina/genética , Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Espermátides/metabolismo , Espermatogénesis/genética , Tubulina (Proteína)/metabolismo , Animales , Carboxipeptidasas/deficiencia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Centriolos/metabolismo , Centriolos/patología , Centriolos/ultraestructura , Citosol/metabolismo , Citosol/ultraestructura , Ácido Glutámico/metabolismo , Humanos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Noqueados , Microtúbulos/patología , Microtúbulos/ultraestructura , Cola del Espermatozoide/metabolismo , Cola del Espermatozoide/patología , Cola del Espermatozoide/ultraestructura , Espermátides/patología , Espermátides/ultraestructura , Tubulina (Proteína)/genética
6.
J Cell Sci ; 130(21): 3749-3763, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28923836

RESUMEN

DNA alkylating agents form the first line of cancer chemotherapy. They not only kill cells but also behave as potential carcinogens. MNU, a DNA methylating agent, is well known to induce mammary tumours in rodents. However, the mechanism of tumorigenesis is not well understood. Our study reports a novel role played by DNA-dependent protein kinase (DNA-PK) in methylation damage-induced transformation using three-dimensional breast acinar cultures. Here, we report that exposure of breast epithelial cells to MNU inhibited polarisation at the basolateral domain, increased dispersal of the Golgi at the apical domain and induced an epithelial-to-mesenchymal transition (EMT)-like phenotype as well as invasion. This altered Golgi phenotype correlated with impaired intracellular trafficking. Inhibition of DNA-PK resulted in almost complete reversal of the altered Golgi phenotype and partial rescue of the polarity defect and EMT-like phenotype. The results confirm that methylation damage-induced activation of DNA-PK is a major mechanism in mediating cellular transformation.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Alquilantes/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Proteína Quinasa Activada por ADN/metabolismo , Células Epiteliales/efectos de los fármacos , Metilnitrosourea/farmacología , Benzaldehídos/farmacología , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ensayo Cometa , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Humanos , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Metilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
7.
J Cell Sci ; 130(8): 1347-1353, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28325758

RESUMEN

Microtubules are key cytoskeletal elements of all eukaryotic cells and are assembled of evolutionarily conserved α-tubulin-ß-tubulin heterodimers. Despite their uniform structure, microtubules fulfill a large diversity of functions. A regulatory mechanism to control the specialization of the microtubule cytoskeleton is the 'tubulin code', which is generated by (i) expression of different α- and ß-tubulin isotypes, and by (ii) post-translational modifications of tubulin. In this Cell Science at a Glance article and the accompanying poster, we provide a comprehensive overview of the molecular components of the tubulin code, and discuss the mechanisms by which these components contribute to the generation of functionally specialized microtubules.


Asunto(s)
Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo , Animales , Movimiento Celular , Humanos , Tubulina (Proteína)/genética
8.
Hippocampus ; 26(10): 1313-27, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27258934

RESUMEN

Although cocaine- and amphetamine-regulated transcript peptide (CART) is detected in several cortical and subcortical areas, its role in higher functions has been largely ignored. We examined the significance of CART in memory formation and tested if the downstream actions of CART involve N-methyl-d-aspartate (NMDA) activated extra-cellular signal-regulated kinase (ERK). Newly formed memory was evaluated using novel object recognition test consisting of familiarization (T1) and choice trials (T2). The choice trials were performed at two time points: 30-min (T230-min ) and 24-h (T224-h ) postacquisition. In choice trial (T230-min ), vehicle control rats explored the novel object for significantly longer duration than the familiar object indicating intact memory formation. However, CART-antibody, U0126 [ERK antagonist, both via intracerebroventricular (icv) or intrahippocampal (ih) route] or MK-801 (NMDA antagonist; intraperitoneal) treated rats spent less time exploring novel objects; CART peptide (icv or ih) was ineffective. During choice trial at T224-h , a significant decrease in novel object exploration time was noticed in vehicle control rats suggesting amnesia. However, treatment with CART, prior to familiarization trial (T1), promoted exploration of the novel object even at T224-h . Pretreatment with U0126 or MK-801 blocked pro-cognitive-like effect of CART suggesting involvement of NMDA-ERK pathway in CART's action. Animals subjected to the object familiarization trial showed a drastic increase in the CART-immunoreactivity in the cells of cornu ammonis 3 and polymorph layer of dentate gyrus, and fibers within ento- (ENT) and peri-rhinal (PRH) cortices. Western blot analysis revealed that CART treatment significantly up-regulated the expression of phospo-ERK1/2 in hippocampus, ENT and PRH. This effect was attenuated following pretreatment with U0126 or MK-801, suggesting the activation of ERK signaling cascade through NMDA receptors. Thus, CART system seems to play an important role in recognition memory and that these effects may be mediated by NMDA receptors-ERK signaling in the ENT/PRH-hippocampal circuit. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Reconocimiento en Psicología/fisiología , Animales , Butadienos/farmacología , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Cognición/efectos de los fármacos , Cognición/fisiología , Maleato de Dizocilpina/farmacología , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/metabolismo , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Proteínas del Tejido Nervioso/administración & dosificación , Neuronas/efectos de los fármacos , Neuronas/fisiología , Pruebas Neuropsicológicas , Nitrilos/farmacología , Corteza Perirrinal/efectos de los fármacos , Corteza Perirrinal/metabolismo , Psicotrópicos/farmacología , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Reconocimiento en Psicología/efectos de los fármacos
9.
BMC Cancer ; 14: 287, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24758542

RESUMEN

BACKGROUND: The DNA damage checkpoint signalling cascade sense damaged DNA and coordinates cell cycle arrest, DNA repair, and/or apoptosis. However, it is still not well understood how the signalling system differentiates between different kinds of DNA damage. N-nitroso-N-ethylurea (NEU), a DNA ethylating agent induces both transversions and transition mutations. METHODS: Immunoblot and comet assays were performed to detect DNA breaks and activation of the canonical checkpoint signalling kinases following NEU damage upto 2 hours. To investigate whether mismatch repair played a role in checkpoint activation, knock-down studies were performed while flow cytometry analysis was done to understand whether the activation of the checkpoint kinases was cell cycle phase specific. Finally, breast epithelial cells were grown as 3-dimensional spheroid cultures to study whether NEU can induce upregulation of vimentin as well as disrupt cell polarity of the breast acini, thus causing transformation of epithelial cells in culture. RESULTS: We report a novel finding that NEU causes activation of major checkpoint signalling kinases, Chk1 and Chk2. This activation is temporally controlled with Chk2 activation preceding Chk1 phosphorylation, and absence of cross talk between the two parallel signalling pathways, ATM and ATR. Damage caused by NEU leads to the temporal formation of both double strand and single strand breaks. Activation of checkpoints following NEU damage is cell cycle phase dependent wherein Chk2 is primarily activated during G2-M phase whilst in S phase, there is immediate Chk1 phosphorylation and delayed Chk2 response. Surprisingly, the mismatch repair system does not play a role in checkpoint activation, at doses and duration of NEU used in the experiments. Interestingly, NEU caused disruption of the well-formed polarised spheroid archithecture and upregulation of vimentin in three-dimensional breast acini cultures of non-malignant breast epithelial cells upon NEU treatment indicating NEU to have the potential to cause early transformation in the cells. CONCLUSION: NEU causes damage in mammalian cells in the form of double strand and single strand breaks that temporally activate the major checkpoint signalling kinases without the occurrence of cross-talk between the pathways. NEU also appear to cause transformation in three-dimensional spheroid cultures.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Quinasa de Punto de Control 2/biosíntesis , Proteínas Quinasas/biosíntesis , Proteínas de la Ataxia Telangiectasia Mutada/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2/metabolismo , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Etilnitrosourea/farmacología , Humanos , Fosforilación , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Front Mol Biosci ; 10: 1252529, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37867557

RESUMEN

Cryogenic electron microscopy (cryo-EM) and electron tomography (cryo-ET) have become a critical tool for studying viral particles. Cryo-EM has enhanced our understanding of viral assembly and replication processes at a molecular resolution. Meanwhile, in situ cryo-ET has been used to investigate how viruses attach to and invade host cells. These advances have significantly contributed to our knowledge of viral biology. Particularly, prompt elucidations of structures of the SARS-CoV-2 spike protein and its variants have directly impacted the development of vaccines and therapeutic measures. This review discusses the progress made by cryo-EM based technologies in comprehending the severe acute respiratory syndrome coronavirus-2 (SARS-Cov-2), the virus responsible for the devastating global COVID-19 pandemic in 2020 with focus on the SARS-CoV-2 spike protein and the mechanisms of the virus entry and replication.

11.
Nat Commun ; 14(1): 620, 2023 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-36739444

RESUMEN

SARS-CoV-2 is a novel coronavirus responsible for the COVID-19 pandemic. Its high pathogenicity is due to SARS-CoV-2 spike protein (S protein) contacting host-cell receptors. A critical hallmark of COVID-19 is the occurrence of coagulopathies. Here, we report the direct observation of the interactions between S protein and platelets. Live imaging shows that the S protein triggers platelets to deform dynamically, in some cases, leading to their irreversible activation. Cellular cryo-electron tomography reveals dense decorations of S protein on the platelet surface, inducing filopodia formation. Hypothesizing that S protein binds to filopodia-inducing integrin receptors, we tested the binding to RGD motif-recognizing platelet integrins and find that S protein recognizes integrin αvß3. Our results infer that the stochastic activation of platelets is due to weak interactions of S protein with integrin, which can attribute to the pathogenesis of COVID-19 and the occurrence of rare but severe coagulopathies.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Plaquetas/metabolismo , Pandemias
12.
J Cell Biol ; 221(4)2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35262630

RESUMEN

Neurons are highly polarized cells forming an intricate network of dendrites and axons. They are shaped by the dynamic reorganization of cytoskeleton components and cellular organelles. Axon branching allows the formation of new paths and increases circuit complexity. However, our understanding of branch formation is sparse due to the lack of direct in-depth observations. Using in situ cellular cryo-electron tomography on primary mouse neurons, we directly visualized the remodeling of organelles and cytoskeleton structures at axon branches. Strikingly, branched areas functioned as hotspots concentrating organelles to support dynamic activities. Unaligned actin filaments assembled at the base of premature branches accompanied by filopodia-like protrusions. Microtubules and ER comigrated into preformed branches to support outgrowth together with accumulating compact, ∼500-nm mitochondria and locally clustered ribosomes. We obtained a roadmap of events supporting the hypothesis of local protein synthesis selectively taking place at axon branches, allowing them to serve as unique control hubs for axon development and downstream neural network formation.


Asunto(s)
Axones , Tomografía con Microscopio Electrónico , Neurogénesis , Actinas , Animales , Axones/metabolismo , Citoesqueleto/metabolismo , Retículo Endoplásmico , Ratones , Microtúbulos/metabolismo , Neurogénesis/fisiología , Biosíntesis de Proteínas
13.
Sci Adv ; 8(10): eabj9229, 2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35275727

RESUMEN

Mutations in the brain-specific ß-tubulin 4A (TUBB4A) gene cause a broad spectrum of diseases, ranging from dystonia (DYT-TUBB4A) to hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC). Currently, the mechanisms of how TUBB4A variants lead to this pleiotropic manifestation remain elusive. Here, we investigated whether TUBB4A mutations causing either DYT-TUBB4A (p.R2G and p.Q424H) or H-ABC (p.R2W and p.D249N) exhibit differential effects at the molecular and cellular levels. Using live-cell imaging of disease-relevant oligodendrocytes and total internal reflection fluorescence microscopy of whole-cell lysates, we observed divergent impact on microtubule polymerization and microtubule integration, partially reflecting the observed pleiotropy. Moreover, in silico simulations demonstrated that the mutants rarely adopted a straight heterodimer conformation in contrast to wild type. In conclusion, for most of the examined variants, we deciphered potential molecular disease mechanisms that may lead to the diverse clinical manifestations and phenotype severity across and within each TUBB4A-related disease.


Asunto(s)
Distonía , Tubulina (Proteína) , Ganglios Basales/metabolismo , Ganglios Basales/patología , Cerebelo/metabolismo , Distonía/genética , Distonía/patología , Humanos , Mutación , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
14.
Nat Cell Biol ; 24(2): 253-267, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35102268

RESUMEN

The microtubule cytoskeleton forms complex macromolecular assemblies with a range of microtubule-associated proteins (MAPs) that have fundamental roles in cell architecture, division and motility. Determining how an individual MAP modulates microtubule behaviour is an important step in understanding the physiological roles of various microtubule assemblies. To characterize how MAPs control microtubule properties and functions, we developed an approach allowing for medium-throughput analyses of MAPs in cell-free conditions using lysates of mammalian cells. Our pipeline allows for quantitative as well as ultrastructural analyses of microtubule-MAP assemblies. Analysing 45 bona fide and potential mammalian MAPs, we uncovered previously unknown activities that lead to distinct and unique microtubule behaviours such as microtubule coiling or hook formation, or liquid-liquid phase separation along the microtubule lattice that initiates microtubule branching. We have thus established a powerful tool for a thorough characterization of a wide range of MAPs and MAP variants, thus opening avenues for the determination of mechanisms underlying their physiological roles and pathological implications.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Imagen Individual de Molécula , Fracciones Subcelulares , Animales , Línea Celular Tumoral , Microscopía por Crioelectrón , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Microscopía por Video , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/ultraestructura , Microtúbulos/genética , Microtúbulos/ultraestructura , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura , Transducción de Señal , Factores de Tiempo , Imagen de Lapso de Tiempo , Tubulina (Proteína)/metabolismo
15.
J Thromb Haemost ; 20(2): 461-469, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34704371

RESUMEN

BACKGROUND: In the panel of genes commonly associated with inherited macrothrombocytopenia, an important fraction encodes key cytoskeletal proteins such as tubulin isotypes, the building blocks of microtubules. Macrothrombocytopenia-causing mutations have been identified in the TUBB1 and TUBA4A genes, emphasizing their importance in the formation of platelets and their marginal band, a unique microtubule ring-like structure that supports the platelet typical disc-shaped morphology. This raised the hypothesis that other tubulin isotypes normally expressed in platelets could play a similar role in their formation. OBJECTIVES: To assess whether tubulin isotype genes other than TUBA4A and TUBB1 could be implicated in inherited macrothrombocytopenia. METHODS: We used high throughput sequencing to screen a cohort of 448 French blood donors with mild thrombocytopenia for mutations in a panel of selected genes known or suspected to be involved in platelet biogenesis. RESULTS: We identified six distinct novel mutations in TUBA8, which encodes the most-divergent α-tubulin, as the causative determinant of macrothrombocytopenia and platelet marginal band defects. Functionally, all TUBA8 mutations were found to fully or partially inhibit the incorporation of the mutated α8-tubulin in the microtubule network. CONCLUSION: This study provides strong support for a key role of multiple tubulin genes in platelet biogenesis by discovering variants in a tubulin gene that was previously not known to be important for platelets.


Asunto(s)
Trombocitopenia , Tubulina (Proteína) , Plaquetas/metabolismo , Humanos , Mutación , Trombocitopenia/genética , Trombocitopenia/metabolismo , Tubulina (Proteína)/genética
16.
bioRxiv ; 2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36451880

RESUMEN

SARS-CoV-2 is a novel coronavirus responsible for the COVID-19 pandemic. Its high pathogenicity is due to SARS-CoV-2 spike protein (S protein) contacting host-cell receptors. A critical hallmark of COVID-19 is the occurrence of coagulopathies. Here, we report the direct observation of the interactions between S protein and platelets. Live imaging showed that the S protein triggers platelets to deform dynamically, in some cases, leading to their irreversible activation. Strikingly, cellular cryo-electron tomography revealed dense decorations of S protein on the platelet surface, inducing filopodia formation. Hypothesizing that S protein binds to filopodia-inducing integrin receptors, we tested the binding to RGD motif-recognizing platelet integrins and found that S protein recognizes integrin α v ß 3 . Our results infer that the stochastic activation of platelets is due to weak interactions of S protein with integrin, which can attribute to the pathogenesis of COVID-19 and the occurrence of rare but severe coagulopathies.

17.
Neurosci Lett ; 746: 135656, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33482309

RESUMEN

Neurodegenerative diseases lead to a progressive demise of neuronal functions that ultimately results in neuronal death. Besides a large variety of molecular pathways that have been linked to the degeneration of neurons, dysfunctions of the microtubule cytoskeleton are common features of many human neurodegenerative disorders. Yet, it is unclear whether microtubule dysfunctions are causative, or mere bystanders in the disease progression. A so-far little explored regulatory mechanism of the microtubule cytoskeleton, the posttranslational modifications of tubulin, emerge as candidate mechanisms involved in neuronal dysfunction, and thus, degeneration. Here we review the role of tubulin polyglutamylation, a prominent modification of neuronal microtubules. We discuss the current understanding of how polyglutamylation controls microtubule functions in healthy neurons, and how deregulation of this modification leads to neurodegeneration in mice and humans.


Asunto(s)
Microtúbulos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Ácido Poliglutámico/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Humanos , Microtúbulos/patología , Enfermedades Neurodegenerativas/patología , Procesamiento Proteico-Postraduccional/fisiología
18.
Front Cell Dev Biol ; 9: 707486, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34540830

RESUMEN

Axon branching is a critical process ensuring a high degree of interconnectivity for neural network formation. As branching occurs at sites distant from the soma, it is necessary that axons have a local system to dynamically control and regulate axonal growth. This machinery depends on the orchestration of cellular functions such as cytoskeleton, subcellular transport, energy production, protein- and membrane synthesis that are adapted for branch formation. Compared to the axon shaft, branching sites show a distinct and dynamic arrangement of cytoskeleton components, endoplasmic reticulum and mitochondria. This review discusses the regulation of axon branching in the context of cytoskeleton and membrane remodeling.

19.
Methods Mol Biol ; 2101: 327-351, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31879912

RESUMEN

Microtubules, as integral part of the eukaryotic cytoskeleton, exert numerous essential functions in cells. A mechanism to control these diverse functions are the posttranslational modifications of tubulin. Despite being known for decades, relatively little insight into the cellular functions of these modifications has been gained so far. The discovery of tubulin-modifying enzymes and a growing number of available knockout mice now allow working with primary cells from those mouse models to address biological functions and molecular mechanisms behind those modifications. However, a number of those mouse models show either lethality or sterility, making it difficult to impossible to obtain a sufficient number of animals for a systematic study with primary cells. Moreover, many of those modifications are controlled by several redundant enzymes, and it is often necessary to knock out several enzymes in parallel to obtain a significant change in a given tubulin modification. Here we describe a method to generate primary cells with combinatorial knockout genotypes using conditional knockout mice. The conditional alleles are converted into knockout in the cultured primary cells by transduction with a lentivirus encoding cre-recombinase. This approach has allowed us to knock out the two main brain deglutamylases in mouse primary neurons, which leads to strongly increased polyglutamylation in these cells. Our method can be applied to measure different cellular processes, such as axonal transport, for which it can be combined with the expression of different fluorescent reporters to label intracellular proteins. Using a panel of conditional knockout mice, our method can further be applied to study the functions of a variety of tubulin modifications that require simultaneous knockout of multiple genes.


Asunto(s)
Técnicas de Silenciamiento del Gen , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Clonación Molecular , Citoesqueleto/genética , Citoesqueleto/metabolismo , Electroforesis en Gel de Poliacrilamida , Expresión Génica , Genes Reporteros , Vectores Genéticos/genética , Humanos , Ratones Noqueados , Células Piramidales/metabolismo , Transducción Genética
20.
Methods Mol Biol ; 2101: 353-370, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31879913

RESUMEN

Axonal transport is a process essential for neuronal function and survival that takes place on the cellular highways-the microtubules. It requires three major components: the microtubules that serve as tracks for the transport, the motor proteins that drive the movement, and the transported cargoes with their adaptor proteins. Axonal transport could be controlled by tubulin posttranslational modifications, which by decorating specific microtubule tracks could determine the specificity of cargo delivery inside neurons. However, it appears that the effects of tubulin modifications on transport can be rather subtle, and might thus be easily overlooked depending on which parameter of the transport process is analyzed. Here we propose an analysis paradigm that allows detecting rather subtle alterations in neuronal transport, as induced for instance by accumulation of posttranslational polyglutamylation. Analyzing mitochondria movements in axons, we found that neither the average speed nor the distance traveled were affected by hyperglutamylation, but we detected an about 50% reduction of the overall motility, suggesting that polyglutamylation controls the efficiency of mitochondria transport in axons. Our protocol can readily be expanded to the analysis of the impact of other tubulin modifications on the transport of a range of different neuronal cargoes.


Asunto(s)
Transporte Axonal , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo , Animales , Axones/metabolismo , Ratones , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Imagen Molecular/métodos , Proteínas Motoras Moleculares/metabolismo , Neuronas/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA