Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.529
Filter
1.
Proc Natl Acad Sci U S A ; 121(38): e2412241121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39254993

ABSTRACT

Dynein is the primary molecular motor responsible for retrograde intracellular transport of a variety of cargoes, performing successive nanometer-sized steps within milliseconds. Due to the limited spatiotemporal precision of established methods for molecular tracking, current knowledge of dynein stepping is essentially limited to slowed-down measurements in vitro. Here, we use MINFLUX fluorophore localization to directly track CRISPR/Cas9-tagged endogenous dynein with nanometer/millisecond precision in living primary neurons. We show that endogenous dynein primarily takes 8 nm steps, including frequent sideways steps but few backward steps. Strikingly, the majority of direction reversals between retrograde and anterograde movement occurred on the time scale of single steps (16 ms), suggesting a rapid regulatory reversal mechanism. Tug-of-war-like behavior during pauses or reversals was unexpectedly rare. By analyzing the dwell time between steps, we concluded that a single rate-limiting process underlies the dynein stepping mechanism, likely arising from just one adenosine 5'-triphosphate hydrolysis event being required during each step. Our study underscores the power of MINFLUX localization to elucidate the spatiotemporal changes underlying protein function in living cells.


Subject(s)
Dyneins , Neurons , Dyneins/metabolism , Neurons/metabolism , Animals , CRISPR-Cas Systems , Adenosine Triphosphate/metabolism , Mice
2.
Hum Genomics ; 18(1): 97, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256880

ABSTRACT

BACKGROUND: Bi-allelic variants in DNAH11 have been identified as causative factors in Primary Ciliary Dyskinesia, leading to abnormal respiratory cilia. Nonetheless, the specific impact of these variants on human sperm flagellar and their involvement in male infertility remain largely unknown. METHODS: A collaborative effort involving two Chinese reproductive centers conducted a study with 975 unrelated infertile men. Whole-exome sequencing was employed for variant screening, and Sanger sequencing confirmed the identified variants. Morphological and ultrastructural analyses of sperm were conducted using Scanning Electron Microscopy and Transmission Electron Microscopy. Western Blot Analysis and Immunofluorescence Analysis were utilized to assess protein levels and localization. ICSI was performed to evaluate its efficacy in achieving favorable pregnancy outcomes for individuals with DNAH11 variants. RESULTS: In this study, we identified seven novel variants in the DNAH11 gene in four asthenoteratozoospermia subjects. These variants led the absence of DNAH11 proteins and ultrastructure defects in sperm flagella, particularly affecting the outer dynein arms (ODAs) and adjacent structures. The levels of ODA protein DNAI2 and axoneme related proteins were down regulated, instead of inner dynein arms (IDA) proteins DNAH1 and DNAH6. Two out of four individuals with DNAH11 variants achieved clinical pregnancies through ICSI. The findings confirm the association between male infertility and bi-allelic deleterious variants in DNAH11, resulting in the aberrant assembly of sperm flagella and contributing to asthenoteratozoospermia. Importantly, ICSI emerges as an effective intervention for overcoming reproductive challenges caused by DNAH11 gene variants.


Subject(s)
Asthenozoospermia , Axonemal Dyneins , Exome Sequencing , Infertility, Male , Humans , Male , Asthenozoospermia/genetics , Asthenozoospermia/pathology , Axonemal Dyneins/genetics , Female , Infertility, Male/genetics , Infertility, Male/pathology , Adult , Sperm Tail/pathology , Sperm Tail/ultrastructure , Sperm Tail/metabolism , Sperm Injections, Intracytoplasmic , Pregnancy , Spermatozoa/ultrastructure , Spermatozoa/pathology , Dyneins/genetics
3.
Ecotoxicol Environ Saf ; 283: 116841, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39128448

ABSTRACT

Lewy body disease (LBD), one of the most common neurodegenerative diseases (NDDs), is characterized by excessive accumulation of α-synuclein (α-syn) in neurons. In recent years, environmental factors such as exposure to herbicides and pesticides have been attributed to the development of this condition. While majority of the studies on neurotoxic effects of paraquat (PQ) have focused on α-syn-mediated neuronal damage in the early stages of α-syn accumulation in neurons, efforts to explore the key target for α-syn degradation are limited. Recent research has suggested that histone deacetylase 6 (HDAC6) might possibly regulate amyloid clearance, and that the metabolism of compounds in neurons is also directly affected by axonal transport in neurons. Dynein predominantly mediates reverse transportation of metabolites and uptake of signal molecules and other compounds at the end of axons, which is conducive to the reuse of cell components. However, the role of interaction of dynein with HDAC6 in metabolites transport is still unclear. Therefore, this study aimed to investigate the role of HDAC6 in α-syn accumulation/clearance in neurons and the associated possible influencing factors. The results revealed that HDAC6 could transport ubiquitinated α-syn, bind to dynein, form an aggresome, and relocate to the center of the microtubule tissue, ultimately reducing abnormal accumulation of α-syn. However, PQ treatment resulted in HDAC6 upregulation, causing abnormal aggregation of α-syn. Taken together, these findings indicated that PQ exposure caused abnormal accumulation of α-syn and decreased effective degradation of α-syn by HDAC6-mediated aggresome-autophagy-lysosome pathway.


Subject(s)
Dyneins , Histone Deacetylase 6 , Paraquat , alpha-Synuclein , Histone Deacetylase 6/metabolism , Paraquat/toxicity , alpha-Synuclein/metabolism , Animals , Dyneins/metabolism , Herbicides/toxicity , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Axonal Transport/drug effects , Mice
4.
Cell Rep ; 43(8): 114649, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159044

ABSTRACT

Each cargo in a cell employs a unique set of motor proteins for its transport. To dissect the roles of each type of motor, we developed optogenetic inhibitors of endogenous kinesin-1, -2, -3 and dynein motors and examined their effect on the transport of early endosomes, late endosomes, and lysosomes. While kinesin-1, -3, and dynein transport vesicles at all stages of endocytosis, kinesin-2 primarily drives late endosomes and lysosomes. Transient optogenetic inhibition of kinesin-1 or dynein causes both early and late endosomes to move more processively by relieving competition with opposing motors. Kinesin-2 and -3 support long-range transport, and optogenetic inhibition reduces the distances that their cargoes move. These results suggest that the directionality of transport is controlled through regulating kinesin-1 and dynein activity. On vesicles transported by several kinesin and dynein motors, modulating the activity of a single type of motor on the cargo is sufficient to direct motility.


Subject(s)
Dyneins , Kinesins , Optogenetics , Kinesins/metabolism , Optogenetics/methods , Dyneins/metabolism , Humans , Animals , Endosomes/metabolism , Lysosomes/metabolism , Biological Transport , HeLa Cells , Endocytosis
5.
ACS Nano ; 18(37): 25893-25905, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39214619

ABSTRACT

After entering host cells by endocytosis, influenza A virus (IAV) is transported along microfilaments and then transported by dynein along microtubules (MTs) to the perinuclear region for genome release. Understanding the mechanisms of dynein-driven transport is significant for a comprehensive understanding of IAV infection. In this work, the roles of dynactin in dynein-driven transport of IAV were quantitatively dissected in situ using quantum dot-based single-virus tracking. It was revealed that dynactin was essential for dynein to transport IAV toward the nucleus. After virus entry, virus-carrying vesicles bound to dynein and dynactin before being delivered to MTs. The attachment of dynein to the vesicles was dependent on dynactin and its subunits, p150Glued and Arp1. Once viruses reached MTs, dynactin-assisted dynein initiates retrograde transport of IAV. Importantly, the retrograde transport of viruses could be initiated at both plus ends (32%) and other regions on MTs (68%). Subsequently, dynactin accompanied and assisted dynein to persistently transport the virus along MTs in the retrograde direction. This study revealed the dynactin-dependent dynein-driven transport process of IAV, enhancing our understanding of IAV infection and providing important insights into the cell's endocytic transport mechanism.


Subject(s)
Dynactin Complex , Dyneins , Influenza A virus , Quantum Dots , Dynactin Complex/metabolism , Dyneins/metabolism , Quantum Dots/chemistry , Quantum Dots/metabolism , Humans , Influenza A virus/metabolism , Biological Transport , Animals , Microtubules/metabolism , Dogs , Madin Darby Canine Kidney Cells , A549 Cells
6.
J Cell Biol ; 223(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39115447

ABSTRACT

Nuclear migration is critical for the proper positioning of neurons in the developing brain. It is known that bidirectional microtubule motors are required for nuclear transport, yet the mechanism of the coordination of opposing motors is still under debate. Using mouse cerebellar granule cells, we demonstrate that Nesprin-2 serves as a nucleus-motor adaptor, coordinating the interplay of kinesin-1 and dynein. Nesprin-2 recruits dynein-dynactin-BicD2 independently of the nearby kinesin-binding LEWD motif. Both motor binding sites are required to rescue nuclear migration defects caused by the loss of function of Nesprin-2. In an intracellular cargo transport assay, the Nesprin-2 fragment encompassing the motor binding sites generates persistent movements toward both microtubule minus and plus ends. Nesprin-2 drives bidirectional cargo movements over a prolonged period along perinuclear microtubules, which advance during the migration of neurons. We propose that Nesprin-2 keeps the nucleus mobile by coordinating opposing motors, enabling continuous nuclear transport along advancing microtubules in migrating cells.


Subject(s)
Cell Nucleus , Dyneins , Kinesins , Microtubule-Associated Proteins , Microtubules , Nerve Tissue Proteins , Neurons , Animals , Microtubules/metabolism , Neurons/metabolism , Kinesins/metabolism , Kinesins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Dyneins/metabolism , Cell Nucleus/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Active Transport, Cell Nucleus , Dynactin Complex/metabolism , Dynactin Complex/genetics , Cell Movement , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cerebellum/metabolism , Cerebellum/cytology , Binding Sites , Humans
7.
Cell Mol Life Sci ; 81(1): 317, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066891

ABSTRACT

Inner dynein arms (IDAs) are formed from a protein complex that is essential for appropriate flagellar bending and beating. IDA defects have previously been linked to the incidence of asthenozoospermia (AZS) and male infertility. The testes-enriched ZMYND12 protein is homologous with an IDA component identified in Chlamydomonas. ZMYND12 deficiency has previously been tied to infertility in males, yet the underlying mechanism remains uncertain. Here, a CRISPR/Cas9 approach was employed to generate Zmynd12 knockout (Zmynd12-/-) mice. These Zmynd12-/- mice exhibited significant male subfertility, reduced sperm motile velocity, and impaired capacitation. Through a combination of co-immunoprecipitation and mass spectrometry, ZMYND12 was found to interact with TTC29 and PRKACA. Decreases in the levels of PRKACA were evident in the sperm of these Zmynd12-/- mice, suggesting that this change may account for the observed drop in male fertility. Moreover, in a cohort of patients with AZS, one patient carrying a ZMYND12 variant was identified, expanding the known AZS-related variant spectrum. Together, these findings demonstrate that ZMYND12 is essential for flagellar beating, capacitation, and male fertility.


Subject(s)
Infertility, Male , Mice, Knockout , Sperm Motility , Animals , Humans , Male , Mice , Asthenozoospermia/genetics , Asthenozoospermia/metabolism , Asthenozoospermia/pathology , CRISPR-Cas Systems , Dyneins/metabolism , Dyneins/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Mice, Inbred C57BL , Sperm Capacitation/genetics , Sperm Motility/genetics , Spermatozoa/metabolism , Contactin 2/genetics , Contactin 2/metabolism
8.
Mol Biol Cell ; 35(9): br16, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39024276

ABSTRACT

The outer dynein arm (ODA) is a large, multimeric protein complex essential for ciliary motility. The composition and assembly of ODA are best characterized in the green algae Chlamydomonas reinhardtii, where individual ODA subunits are synthesized and preassembled into a mature complex in the cytosol prior to ciliary import. The single-cellular parasite Trypanosoma brucei contains a motile flagellum essential for cell locomotion and pathogenesis. Similar to human motile cilia, T. brucei flagellum contains a two-headed ODA complex arranged at 24 nm intervals along the axonemal microtubule doublets. The subunit composition and the preassembly of the ODA complex in T. brucei, however, have not been investigated. In this study, we affinity-purified the ODA complex from T. brucei cytoplasmic extract. Proteomic analyses revealed the presence of two heavy chains (ODAα and ODAß), two intermediate chains (IC1and IC2) and several light chains. We showed that both heavy chains and both intermediate chains are indispensable for flagellar ODA assembly. Our study also provided biochemical evidence supporting the presence of a cytoplasmic, preassembly pathway for T. brucei ODA.


Subject(s)
Axoneme , Cytoplasm , Dyneins , Flagella , Protozoan Proteins , Trypanosoma brucei brucei , Trypanosoma brucei brucei/metabolism , Flagella/metabolism , Cytoplasm/metabolism , Axoneme/metabolism , Dyneins/metabolism , Protozoan Proteins/metabolism , Microtubules/metabolism , Proteomics/methods , Cilia/metabolism
9.
Mol Biol Cell ; 35(9): ar116, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39024292

ABSTRACT

Ninein (Nin) is a microtubule (MT) anchor at the subdistal appendages of mother centrioles and the pericentriolar material (PCM) of centrosomes that also functions to organize MTs at noncentrosomal MT-organizing centers (ncMTOCs). In humans, the NIN gene is mutated in Seckel syndrome, an inherited developmental disorder. Here, we dissect the protein domains involved in Nin's localization and interactions with dynein and ensconsin (ens/MAP7) and show that the association with ens cooperatively regulates MT assembly in Drosophila fat body cells. We define domains of Nin responsible for its localization to the ncMTOC on the fat body cell nuclear surface, localization within the nucleus, and association with Dynein light intermediate chain (Dlic) and ens, respectively. We show that Nin's association with ens synergistically regulates MT assembly. Together, these findings reveal novel features of Nin function and its regulation of a ncMTOC.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Dyneins , Microtubule-Associated Proteins , Microtubule-Organizing Center , Microtubules , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Animals , Dyneins/metabolism , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Drosophila melanogaster/metabolism , Nuclear Proteins/metabolism , Centrosome/metabolism , Protein Domains , Humans , Fat Body/metabolism , Drosophila/metabolism , Cell Nucleus/metabolism , Centrioles/metabolism , Protein Binding , Homeodomain Proteins
10.
Cells ; 13(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39056782

ABSTRACT

Disease-causing bi-allelic DNA variants in CCDC39 and CCDC40 are frequent causes of the hereditary disorder of primary ciliary dyskinesia (PCD). The encoded proteins form a molecular ruler complex, crucial for maintaining the 96 nm repeat units along the ciliary axonemes. Defects of those proteins cause a stiff, rapid, and flickery ciliary beating pattern, recurrent respiratory infections, axonemal disorganization, and abnormal assembly of GAS8, CCDC39, and DNALI1. We performed molecular characterization of the defects in the 96 nm axonemal ruler due to disease-causing variants in CCDC39 and CCDC40 and analyzed the effect on additional axonemal components. We identified a cohort of 51 individuals with disease-causing variants in CCDC39 and CCDC40 via next-generation sequencing techniques and demonstrated that the IDA heavy chains DNAH1, DNAH6, and DNAH7 are conspicuously absent within the respiratory ciliary axonemes by immunofluorescence analyses. Hence, we show for the first time that the centrin2 (CETN2) containing IDAs are also affected. These findings underscore the crucial role of CCDC39 and CCDC40 in the assembly and function of IDAs in human respiratory cilia. Thus, our data improve the diagnostics of axonemal ruler defects by further characterizing the associated molecular IDA defects.


Subject(s)
Axoneme , Humans , Male , Axonemal Dyneins/metabolism , Axonemal Dyneins/genetics , Axoneme/metabolism , Cilia/metabolism , Cilia/pathology , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/metabolism , Ciliary Motility Disorders/pathology , Cytoskeletal Proteins , Dyneins/metabolism , Dyneins/genetics , Mutation/genetics , Proteins
11.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007638

ABSTRACT

Vertebrate motile cilia are classified as (9+2) or (9+0), based on the presence or absence of the central pair apparatus, respectively. Cryogenic electron microscopy analyses of (9+2) cilia have uncovered an elaborate axonemal protein composition. The extent to which these features are conserved in (9+0) cilia remains unclear. CFAP53, a key axonemal filamentous microtubule inner protein (fMIP) and a centriolar satellites component, is essential for motility of (9+0), but not (9+2) cilia. Here, we show that in (9+2) cilia, CFAP53 functions redundantly with a paralogous fMIP, MNS1. MNS1 localises to ciliary axonemes, and combined loss of both proteins in zebrafish and mice caused severe outer dynein arm loss from (9+2) cilia, significantly affecting their motility. Using immunoprecipitation, we demonstrate that, whereas MNS1 can associate with itself and CFAP53, CFAP53 is unable to self-associate. We also show that additional axonemal dynein-interacting proteins, two outer dynein arm docking (ODAD) complex members, show differential localisation between types of motile cilia. Together, our findings clarify how paralogous fMIPs, CFAP53 and MNS1, function in regulating (9+2) versus (9+0) cilia motility, and further emphasise extensive structural diversity among these organelles.


Subject(s)
Axoneme , Cilia , Zebrafish , Animals , Cilia/metabolism , Cilia/ultrastructure , Zebrafish/metabolism , Mice , Axoneme/metabolism , Axoneme/ultrastructure , Axonemal Dyneins/metabolism , Axonemal Dyneins/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Dyneins/metabolism
12.
Nat Commun ; 15(1): 6311, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060258

ABSTRACT

Respiratory syncytial virus (RSV) hijacks cholesterol or autophagy pathways to facilitate optimal replication. However, our understanding of the associated molecular mechanisms remains limited. Here, we show that RSV infection blocks cholesterol transport from lysosomes to the endoplasmic reticulum by downregulating the activity of lysosomal acid lipase, activates the SREBP2-LDLR axis, and promotes uptake and accumulation of exogenous cholesterol in lysosomes. High cholesterol levels impair the VAP-A-binding activity of ORP1L and promote the recruitment of dynein-dynactin, PLEKHM1, or HOPS VPS39 to Rab7-RILP, thereby facilitating minus-end transport of autophagosomes and autolysosome formation. Acidification inhibition and dysfunction of cholesterol-rich lysosomes impair autophagy flux by inhibiting autolysosome degradation, which promotes the accumulation of RSV fusion protein. RSV-F storage is nearly abolished after cholesterol depletion or knockdown of LDLR. Most importantly, the knockout of LDLR effectively inhibits RSV infection in vivo. These findings elucidate the molecular mechanism of how RSV co-regulates lysosomal cholesterol reprogramming and autophagy and reveal LDLR as a novel target for anti-RSV drug development.


Subject(s)
Autophagy , Cholesterol , Lysosomes , Receptors, LDL , Respiratory Syncytial Virus Infections , Vesicular Transport Proteins , Virus Replication , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Lysosomes/metabolism , Cholesterol/metabolism , Humans , Animals , Receptors, LDL/metabolism , Receptors, LDL/genetics , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Mice , Dynactin Complex/metabolism , Endoplasmic Reticulum/metabolism , Dyneins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Respiratory Syncytial Virus, Human/physiology , Autophagosomes/metabolism , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/genetics , HeLa Cells , A549 Cells
13.
Cell Death Dis ; 15(7): 499, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997255

ABSTRACT

Dynein complexes are large, multi-unit assemblies involved in many biological processes via their critical roles in protein transport and axoneme motility. Using next-generation sequencing of infertile men presenting with low or no sperm in their ejaculates, we identified damaging variants in the dynein-related gene AXDND1. We thus hypothesised that AXDND1 is a critical regulator of male fertility. To test this hypothesis, we produced a knockout mouse model. Axdnd1-/- males were sterile at all ages but presented with an evolving testis phenotype wherein they could undergo one round of histologically replete spermatogenesis followed by a rapid depletion of the seminiferous epithelium. Marker experiments identified a role for AXDND1 in maintaining the balance between differentiation-committed and self-renewing spermatogonial populations, resulting in disproportionate production of differentiating cells in the absence of AXDND1 and increased sperm production during initial spermatogenic waves. Moreover, long-term spermatogonial maintenance in the Axdnd1 knockout was compromised, ultimately leading to catastrophic germ cell loss, destruction of blood-testis barrier integrity and immune cell infiltration. In addition, sperm produced during the first wave of spermatogenesis were immotile due to abnormal axoneme structure, including the presence of ectopic vesicles and abnormalities in outer dense fibres and microtubule doublet structures. Sperm output was additionally compromised by a severe spermiation defect and abnormal sperm individualisation. Collectively these data identify AXDND1 as an atypical dynein complex-related protein with a role in protein/vesicle transport of relevance to spermatogonial function and sperm tail formation in mice and humans. This study underscores the importance of studying the consequences of gene loss-of-function on both the establishment and maintenance of male fertility.


Subject(s)
Mice, Knockout , Sperm Tail , Spermatogenesis , Spermatogonia , Animals , Humans , Male , Mice , Cell Differentiation , Dyneins/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Mice, Inbred C57BL , Sperm Tail/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Testis/metabolism , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism
14.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38949648

ABSTRACT

The diverse roles of the dynein motor in shaping microtubule networks and cargo transport complicate in vivo analysis of its functions significantly. To address this issue, we have generated a series of missense mutations in Drosophila Dynein heavy chain. We show that mutations associated with human neurological disease cause a range of defects, including impaired cargo trafficking in neurons. We also describe a novel microtubule-binding domain mutation that specifically blocks the metaphase-anaphase transition during mitosis in the embryo. This effect is independent from dynein's canonical role in silencing the spindle assembly checkpoint. Optical trapping of purified dynein complexes reveals that this mutation only compromises motor performance under load, a finding rationalized by the results of all-atom molecular dynamics simulations. We propose that dynein has a novel function in anaphase progression that depends on it operating in a specific load regime. More broadly, our work illustrates how in vivo functions of motors can be dissected by manipulating their mechanical properties.


Subject(s)
Anaphase , Drosophila Proteins , Drosophila melanogaster , Dyneins , Microtubules , Animals , Dyneins/metabolism , Dyneins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Microtubules/metabolism , Microtubules/genetics , Molecular Dynamics Simulation , Mutation/genetics , Spindle Apparatus/metabolism , Spindle Apparatus/genetics , Humans , Mutation, Missense
15.
Biophys Rep (N Y) ; 4(3): 100171, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-38996867

ABSTRACT

A common type of cytoskeletal morphology involves multiple microtubules converging with their minus ends at the microtubule organizing center (MTOC). The cargo-motor complex will experience ballistic transport when bound to microtubules or diffusive transport when unbound. This machinery allows for sequestering and subsequent dispersal of dynein-transported cargo. The general principles governing dynamics, efficiency, and tunability of such transport in the MTOC vicinity are not fully understood. To address this, we develop a one-dimensional model that includes advective transport toward an attractor (such as the MTOC) and diffusive transport that allows particles to reach absorbing boundaries (such as cellular membranes). We calculated the mean first passage time (MFPT) for cargo to reach the boundaries as a measure of the effectiveness of sequestering (large MFPT) and diffusive dispersal (low MFPT). We show that the MFPT experiences a dramatic growth, transitioning from a low to high MFPT regime (dispersal to sequestering) over a window of cargo on-/off-rates that is close to in vivo values. Furthermore, increasing either the on-rate (attachment) or off-rate (detachment) can result in optimal dispersal when the attractor is placed asymmetrically. Finally, we also describe a regime of rare events where the MFPT scales exponentially with motor velocity and the escape location becomes exponentially sensitive to the attractor positioning. Our results suggest that structures such as the MTOC allow for the sensitive control of the spatial and temporal features of transport and corresponding function under physiological conditions.


Subject(s)
Microtubules , Microtubules/metabolism , Biological Transport , Models, Biological , Microtubule-Organizing Center/metabolism , Dyneins/metabolism
16.
J Cell Sci ; 137(13)2024 07 01.
Article in English | MEDLINE | ID: mdl-38853670

ABSTRACT

Intraflagellar transport (IFT) is required for ciliary assembly. The IFT machinery comprises the IFT motors kinesin-2 and IFT dynein plus IFT-A and IFT-B complexes, which assemble into IFT trains in cilia. To gain mechanistic understanding of IFT and ciliary assembly, here, we performed an absolute quantification of IFT machinery in Chlamydomonas reinhardtii cilium. There are ∼756, ∼532, ∼276 and ∼350 molecules of IFT-B, IFT-A, IFT dynein and kinesin-2, respectively, per cilium. The amount of IFT-B is sufficient to sustain rapid ciliary growth in terms of tubulin delivery. The stoichiometric ratio of IFT-B:IFT-A:dynein is ∼3:2:1 whereas the IFT-B:IFT-A ratio in an IFT dynein mutant is 2:1, suggesting that there is a plastic interaction between IFT-A and IFT-B that can be influenced by IFT dynein. Considering diffusion of kinesin-2 during retrograde IFT, it is estimated that one kinesin-2 molecule drives eight molecules of IFT-B during anterograde IFT. These data provide new insights into the assembly of IFT trains and ciliary assembly.


Subject(s)
Chlamydomonas reinhardtii , Cilia , Dyneins , Flagella , Kinesins , Proteomics , Cilia/metabolism , Chlamydomonas reinhardtii/metabolism , Chlamydomonas reinhardtii/genetics , Proteomics/methods , Kinesins/metabolism , Kinesins/genetics , Dyneins/metabolism , Flagella/metabolism , Biological Transport
17.
Mol Biol Cell ; 35(8): ar106, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38865178

ABSTRACT

Outer dynein arms (ODAs) are responsible for ciliary beating in eukaryotes. They are assembled in the cytoplasm and shipped by intraflagellar transport (IFT) before attachment to microtubule doublets via the docking complex. The LRRC56 protein has been proposed to contribute to ODAs maturation. Mutations or deletion of the LRRC56 gene lead to reduced ciliary motility in all species investigated so far, but with variable impact on dynein arm presence. Here, we investigated the role of LRRC56 in the protist Trypanosoma brucei, where its absence results in distal loss of ODAs, mostly in growing flagella. We show that LRRC56 is a transient cargo of IFT trains during flagellum construction and surprisingly, is required for efficient attachment of a subset of docking complex proteins present in the distal portion of the organelle. This relation is interdependent since the knockdown of the distal docking complex prevents LRRC56's association with the flagellum. Intriguingly, lrrc56-/- cells display shorter flagella whose maturation is delayed. Inhibition of cell division compensates for the distal ODAs absence thanks to the redistribution of the proximal docking complex, restoring ODAs attachment but not the flagellum length phenotype. This work reveals an unexpected connection between LRRC56 and the docking complex.


Subject(s)
Dyneins , Flagella , Protozoan Proteins , Trypanosoma brucei brucei , Trypanosoma brucei brucei/metabolism , Flagella/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Dyneins/metabolism , Microtubules/metabolism , Cilia/metabolism , Biological Transport/physiology , Axoneme/metabolism
18.
Dev Cell ; 59(18): 2429-2442.e4, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-38866013

ABSTRACT

The forces that orient the spindle in human cells remain poorly understood due to a lack of direct mechanical measurements in mammalian systems. We use magnetic tweezers to measure the force on human mitotic spindles. Combining the spindle's measured resistance to rotation, the speed at which it rotates after laser ablating astral microtubules, and estimates of the number of ablated microtubules reveals that each microtubule contacting the cell cortex is subject to ∼5 pN of pulling force, suggesting that each is pulled on by an individual dynein motor. We find that the concentration of dynein at the cell cortex and extent of dynein clustering are key determinants of the spindle's resistance to rotation, with little contribution from cytoplasmic viscosity, which we explain using a biophysically based mathematical model. This work reveals how pulling forces on astral microtubules determine the mechanics of spindle orientation and demonstrates the central role of cortical dynein clustering.


Subject(s)
Dyneins , Microtubules , Mitosis , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Microtubules/metabolism , Dyneins/metabolism , Mitosis/physiology
19.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38869473

ABSTRACT

At each cell division, nanometer-scale motors and microtubules give rise to the micron-scale spindle. Many mitotic motors step helically around microtubules in vitro, and most are predicted to twist the spindle in a left-handed direction. However, the human spindle exhibits only slight global twist, raising the question of how these molecular torques are balanced. Here, we find that anaphase spindles in the epithelial cell line MCF10A have a high baseline twist, and we identify factors that both increase and decrease this twist. The midzone motors KIF4A and MKLP1 are together required for left-handed twist at anaphase, and we show that KIF4A generates left-handed torque in vitro. The actin cytoskeleton also contributes to left-handed twist, but dynein and its cortical recruitment factor LGN counteract it. Together, our work demonstrates that force generators regulate twist in opposite directions from both within and outside the spindle, preventing strong spindle twist during chromosome segregation.


Subject(s)
Anaphase , Kinesins , Microtubules , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Kinesins/metabolism , Kinesins/genetics , Microtubules/metabolism , Dyneins/metabolism , Dyneins/genetics , Torque , Chromosome Segregation , Actin Cytoskeleton/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
20.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38935075

ABSTRACT

Regulated cell shape change requires the induction of cortical cytoskeletal domains. Often, local changes to plasma membrane (PM) topography are involved. Centrosomes organize cortical domains and can affect PM topography by locally pulling the PM inward. Are these centrosome effects coupled? At the syncytial Drosophila embryo cortex, centrosome-induced actin caps grow into dome-like compartments for mitoses. We found the nascent cap to be a collection of PM folds and tubules formed over the astral centrosomal MT array. The localized infoldings require centrosome and dynein activities, and myosin-based surface tension prevents them elsewhere. Centrosome-engaged PM infoldings become specifically enriched with an Arp2/3 induction pathway. Arp2/3 actin network growth between the infoldings counterbalances centrosomal pulling forces and disperses the folds for actin cap expansion. Abnormal domain topography with either centrosome or Arp2/3 disruption correlates with decreased exocytic vesicle association. Together, our data implicate centrosome-organized PM infoldings in coordinating Arp2/3 network growth and exocytosis for cortical domain assembly.


Subject(s)
Actin-Related Protein 2-3 Complex , Actins , Cell Membrane , Centrosome , Drosophila Proteins , Drosophila melanogaster , Animals , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actins/metabolism , Cell Membrane/metabolism , Centrosome/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Dyneins/metabolism , Exocytosis , Microtubules/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL