Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.401
Filter
1.
Open Biol ; 14(9): 240036, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39255847

ABSTRACT

Family with sequence similarity 161 (Fam161) is an ancient family of microtubule-binding proteins located at the centriole and cilium transition zone (TZ) lumen that exhibit rapid evolution in mice. However, their adaptive role is unclear. Here, we used flies to gain insight into their cell type-specific adaptations. Fam161 is the sole orthologue of FAM161A and FAM161B found in flies. Mutating Fam161 results in reduced male reproduction and abnormal geotaxis behaviour. Fam161 localizes to sensory neuron centrioles and their specialized TZ (the connecting cilium) in a cell type-specific manner, sometimes labelling only the centrioles, sometimes labelling the centrioles and cilium TZ and sometimes labelling the TZ with varying lengths that are longer than other TZ proteins, defining a new ciliary compartment, the extra distal TZ. These findings suggest that Fam161 is an essential centriole and TZ protein with a unique cell type-specific localization in fruit flies that can produce cell type-specific adaptations.


Subject(s)
Centrioles , Cilia , Drosophila Proteins , Animals , Centrioles/metabolism , Cilia/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Male , Drosophila melanogaster/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Organ Specificity
2.
J Cell Biol ; 223(12)2024 Dec 02.
Article in English | MEDLINE | ID: mdl-39316454

ABSTRACT

Subcortical heterotopia is a cortical malformation associated with epilepsy, intellectual disability, and an excessive number of cortical neurons in the white matter. Echinoderm microtubule-associated protein like 1 (EML1) mutations lead to subcortical heterotopia, associated with abnormal radial glia positioning in the cortical wall, prior to malformation onset. This perturbed distribution of proliferative cells is likely to be a critical event for heterotopia formation; however, the underlying mechanisms remain unexplained. This study aimed to decipher the early cellular alterations leading to abnormal radial glia. In a forebrain conditional Eml1 mutant model and human patient cells, primary cilia and centrosomes are altered. Microtubule dynamics and cell cycle kinetics are also abnormal in mouse mutant radial glia. By rescuing microtubule formation in Eml1 mutant embryonic brains, abnormal radial glia delamination and heterotopia volume were significantly reduced. Thus, our new model of subcortical heterotopia reveals the causal link between Eml1's function in microtubule regulation and cell position, both critical for correct cortical development.


Subject(s)
Centrosome , Microtubule-Associated Proteins , Microtubules , Prosencephalon , Animals , Centrosome/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Prosencephalon/metabolism , Prosencephalon/pathology , Prosencephalon/embryology , Microtubules/metabolism , Mice , Cilia/metabolism , Cilia/pathology , Mutation/genetics , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Cell Cycle/genetics
3.
Zhen Ci Yan Jiu ; 49(8): 821-828, 2024 Aug 25.
Article in English, Chinese | MEDLINE | ID: mdl-39318311

ABSTRACT

OBJECTIVES: To observe the effects of electroacupuncture (EA) on the morphological changes of intervertebral disc tissues, apoptosis of nucleus pulposus cells, and the protein expression of Unc-51 like autophagy-activated kinase 1 (ULK1), homologous series of yeast Atg6 (Beclin1), and light chain protease complication 3 type (LC3) in nucleus pulposus tissue of cervical spondylosis rabbits, so as to explore the role of cellular autophagy in EA treatment of cervical spondylosis. METHODS: A total of 24 New Zealand white rabbits were randomly divided into blank, model and EA groups, with 8 rabbits in each group. In the EA group, both sides of the cervical (C)3-C6 "Jiaji" (EX-B2) were stimulated by EA (2 Hz/100 Hz, 1 mA) for 25 min, once daily for 5 days in a course, with a 2-day interval between courses, totaling 4 treatment courses. X-ray was used to assess cervical spine radiographic changes and evaluate radiographic scores;transmission electron microscopy was used to observe ultrastructural changes in nucleus pulposus cells;HE staining was used to observe morphological changes of intervertebral disc tissues and conduct pathological scoring;TUNEL staining was used to observe apoptosis rate of nucleus pulposus cells;Western blot was performed to detect protein expression levels of ULK1, Beclin1, and LC3 in nucleus pulposus tissue. RESULTS: Compared with the blank group, rabbits in the model group showed significantly higher cervical spine radiographic scores (P<0.01), higher pathological scores of intervertebral disc tissues (P<0.05), increased apoptosis rate of nucleus pulposus cells (P<0.01), and decreased expression levels of ULK1, Beclin1, and LC3Ⅱ proteins in nucleus pulposus tissue (P<0.05). Compared with the model group, the EA group showed significantly lower pathological scores of intervertebral discs (P<0.05), lower apoptosis rate of nucleus pulposus cells (P<0.01), and higher protein expression levels of ULK1, Beclin1, and LC3Ⅱ in nucleus pulposus tissue (P<0.01). Rabbits in the blank control group exhibited generally normal organelle structures in nucleus pulposus tissues with few autophagic vacuoles, indicative of early stages of autophagy;while those in the model group showed disrupted organelle structures with cytoplasmic condensation and those in the EA group exhibited autophagosomes with double-membrane structures in nucleus pulposus tissues. CONCLUSIONS: EA promotes the expression of ULK1, Beclin1, and LC3Ⅱ proteins in nucleus pulposus tissues, reduces apoptosis of nucleus pulposus cells, and improves intervertebral disc degeneration.


Subject(s)
Acupuncture Points , Autophagy , Electroacupuncture , Nucleus Pulposus , Spondylosis , Animals , Rabbits , Nucleus Pulposus/metabolism , Spondylosis/therapy , Spondylosis/metabolism , Spondylosis/genetics , Humans , Male , Apoptosis , Beclin-1/metabolism , Beclin-1/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Female , Cervical Vertebrae/metabolism , Intervertebral Disc/metabolism
4.
Proc Natl Acad Sci U S A ; 121(37): e2402817121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39236246

ABSTRACT

Autophagy of glycogen (glycophagy) is crucial for the maintenance of cellular glucose homeostasis and physiology in mammals. STBD1 can serve as an autophagy receptor to mediate glycophagy by specifically recognizing glycogen and relevant key autophagic factors, but with poorly understood mechanisms. Here, we systematically characterize the interactions of STBD1 with glycogen and related saccharides, and determine the crystal structure of the STBD1 CBM20 domain with maltotetraose, uncovering a unique binding mode involving two different oligosaccharide-binding sites adopted by STBD1 CBM20 for recognizing glycogen. In addition, we demonstrate that the LC3-interacting region (LIR) motif of STBD1 can selectively bind to six mammalian ATG8 family members. We elucidate the detailed molecular mechanism underlying the selective interactions of STBD1 with ATG8 family proteins by solving the STBD1 LIR/GABARAPL1 complex structure. Importantly, our cell-based assays reveal that both the STBD1 LIR/GABARAPL1 interaction and the intact two oligosaccharide binding sites of STBD1 CBM20 are essential for the effective association of STBD1, GABARAPL1, and glycogen in cells. Finally, through mass spectrometry, biochemical, and structural modeling analyses, we unveil that STBD1 can directly bind to the Claw domain of RB1CC1 through its LIR, thereby recruiting the key autophagy initiation factor RB1CC1. In all, our findings provide mechanistic insights into the recognitions of glycogen, ATG8 family proteins, and RB1CC1 by STBD1 and shed light on the potential working mechanism of STBD1-mediated glycophagy.


Subject(s)
Autophagy-Related Protein 8 Family , Autophagy , Glycogen , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Autophagy/physiology , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Protein 8 Family/chemistry , Binding Sites , Crystallography, X-Ray , Glycogen/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , Protein Binding
5.
PeerJ ; 12: e18037, 2024.
Article in English | MEDLINE | ID: mdl-39238827

ABSTRACT

Background: This study investigated the genetic characteristics of five Chinese families with keratoconus (KC). Methods: In the five families affected by KC, medical records, clinical observations, and blood samples were collected from all individuals. All KC family members (n = 20) underwent both whole exome sequencing of genomic DNA and Sanger sequencing to confirm the variants. Online software was utilized to analyze all variants, and the online server I-TASSER was employed for in silico predictions of the three-dimensional protein structures of the variants. The newly discovered variants and single nucleotide polymorphisms were further examined in 322 sporadic KC patients. Results: The Pentacam tomographic composite index in those affected first-degree family members of the probands showed a pathological change. Five new variants were detected in the five probands and other affected members in their families: a heterozygous missense variant g.19043832C>T (p.Ser145Asn) in the homer scaffolding protein 3 (HOMER3) gene; a heterozygous missense variant g.99452113G>A (p.Gly483Arg) in the insulin-like growth factor 1 receptor (IGF1R) gene; a heterozygous missense variant g.55118280G>T (p.Trp843Leu) in the echinoderm microtubule-associated protein like 6 (EML6) gene; a heterozygous frameshift variant c. 1226_1227del (p.Gln410Glufs*17) in the DOP1 leucine zipper-like protein B (DOP1B) gene; and a heterozygous splice-site variant c.7776+2T>A in the neurobeachin-like protein 2 (NBEAL2) gene. These variations were predicted to be potentially pathogenic and associated with KC. Conclusion: Five novel variants in HOMER3, IGF1R, EML6, DOP1B, and NBEAL2 genes were identified in this study and may be associated with the pathogenesis of KC. This study provides new information about the gene variants and their protein changes in KC patients. The findings should be explored further and could potentially be applied to the early diagnosis of KC before clinical onset.


Subject(s)
Keratoconus , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult , China , East Asian People/genetics , Exome Sequencing , Genetic Predisposition to Disease/genetics , Homeodomain Proteins/genetics , Keratoconus/genetics , Microtubule-Associated Proteins/genetics , Mutation, Missense , Pedigree , Polymorphism, Single Nucleotide , Receptor, IGF Type 1/genetics , Child
6.
Science ; 385(6715): 1366-1375, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298589

ABSTRACT

Faithful chromosome segregation requires biorientation, where the pair of kinetochores on the chromosome establish bipolar microtubule attachment. The integrity of the kinetochore, a macromolecular complex built on centromeric DNA, is required for biorientation, but components sufficient for biorientation remain unknown. Here, we show that tethering the outer kinetochore heterodimer NDC80-NUF2 to the surface of apolar microbeads establishes their biorientation-like state in mouse cells. NDC80-NUF2 microbeads align at the spindle equator and self-correct alignment errors. The alignment is associated with stable bipolar microtubule attachment and is independent of the outer kinetochore proteins SPC24-SPC25, KNL1, the Mis12 complex, inner kinetochore proteins, and Aurora. Larger microbeads align more rapidly, suggesting a size-dependent biorientation mechanism. This study demonstrates a biohybrid kinetochore design for synthetic biorientation of microscale particles in cells.


Subject(s)
Cell Cycle Proteins , Chromosome Segregation , Kinetochores , Microspheres , Microtubule-Associated Proteins , Microtubules , Spindle Apparatus , Animals , Mice , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Spindle Apparatus/metabolism
7.
Acta Neuropathol ; 148(1): 45, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39305312

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron disease with a mean survival time of three years. The 97% of the cases have TDP-43 nuclear depletion and cytoplasmic aggregation in motor neurons. TDP-43 prevents non-conserved cryptic exon splicing in certain genes, maintaining transcript stability, including ATG4B, which is crucial for autophagosome maturation and Microtubule-associated proteins 1A/1B light chain 3B (LC3B) homeostasis. In ALS mice (G93A), Atg4b depletion worsens survival rates and autophagy function. For the first time, we observed an elevation of LC3ylation in the CNS of both ALS patients and atg4b-/- mouse spinal cords. Furthermore, LC3ylation modulates the distribution of ATG3 across membrane compartments. Antisense oligonucleotides (ASOs) targeting cryptic exon restore ATG4B mRNA in TARDBP knockdown cells. We further developed multi-target ASOs targeting TDP-43 binding sequences for a broader effect. Importantly, our ASO based in peptide-PMO conjugates show brain distribution post-IV administration, offering a non-invasive ASO-based treatment avenue for neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Autophagy-Related Proteins , Cysteine Endopeptidases , DNA-Binding Proteins , Microtubule-Associated Proteins , Animals , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Male , Spinal Cord/metabolism , Spinal Cord/pathology , Autophagy/physiology , Mice, Knockout , RNA Splicing/genetics , Female , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Oligonucleotides, Antisense/pharmacology
8.
Nat Commun ; 15(1): 7967, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261498

ABSTRACT

Microtubule-based vesicle trafficking usually relies upon kinesin and dynein motors and few reports describe microtubule polymerisation driving directional vesicle trafficking. Here we show that Arabidopsis END BINDING1b (EB1b), a microtubule plus-end binding protein, directly interacts with SYP121, a SNARE protein that mediates the trafficking of the K+ channel KAT1 and its distribution to the plasma membrane (PM) in Arabidopsis guard cells. Knockout of AtEB1b and its homologous proteins results in a modest but significant change in the distribution of KAT1 and SYP121 in guard cells and consequently delays light-induced stomatal opening. Live-cell imaging reveals that a portion of SYP121-associated endomembrane compartments co-localise with AtEB1b at the growing ends of microtubules, trafficking along with the growth of microtubules for targeting to the PM. Our study reveals a mechanism of vesicle trafficking driven by microtubule growth, which is involved in the redistribution of PM proteins to modulate guard cell movement.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Membrane , Microtubule-Associated Proteins , Microtubules , Plant Stomata , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Microtubules/metabolism , Plant Stomata/metabolism , Plant Stomata/physiology , Cell Membrane/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Protein Transport , Katanin/metabolism , Katanin/genetics , Cell Movement , Cell Cycle Proteins
9.
PLoS Genet ; 20(9): e1011373, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226307

ABSTRACT

Although centrosomes help organize spindles in most cell types, oocytes of most species lack these structures. During acentrosomal spindle assembly in C. elegans oocytes, microtubule minus ends are sorted outwards away from the chromosomes where they form poles, but then these outward forces must be balanced to form a stable bipolar structure. Simultaneously, microtubule dynamics must be precisely controlled to maintain spindle length and organization. How forces and dynamics are tuned to create a stable bipolar structure is poorly understood. Here, we have gained insight into this question through studies of ZYG-8, a conserved doublecortin-family kinase; the mammalian homolog of this microtubule-associated protein is upregulated in many cancers and has been implicated in cell division, but the mechanisms by which it functions are poorly understood. We found that ZYG-8 depletion from oocytes resulted in overelongated spindles with pole and midspindle defects. Importantly, experiments with monopolar spindles revealed that ZYG-8 depletion led to excess outward forces within the spindle and suggested a potential role for this protein in regulating the force-generating motor BMK-1/kinesin-5. Further, we found that ZYG-8 is also required for proper microtubule dynamics within the oocyte spindle and that kinase activity is required for its function during both meiosis and mitosis. Altogether, our findings reveal new roles for ZYG-8 in oocytes and provide insights into how acentrosomal spindles are stabilized to promote faithful meiosis.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Microtubules , Oocytes , Spindle Apparatus , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Microtubules/metabolism , Microtubules/genetics , Spindle Apparatus/metabolism , Spindle Apparatus/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Oocytes/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
10.
Zool Res ; 45(5): 1073-1087, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39245651

ABSTRACT

Infertility represents a significant health concern, with sperm quantity and quality being crucial determinants of male fertility. Oligoasthenoteratozoospermia (OAT) is characterized by reduced sperm motility, lower sperm concentration, and morphological abnormalities in sperm heads and flagella. Although variants in several genes have been implicated in OAT, its genetic etiologies and pathogenetic mechanisms remain inadequately understood. In this study, we identified a homozygous nonsense mutation (c.916C>T, p.Arg306*) in the coiled-coil domain containing 146 ( CCDC146) gene in an infertile male patient with OAT. This mutation resulted in the production of a truncated CCDC146 protein (amino acids 1-305), retaining only two out of five coiled-coil domains. To validate the pathogenicity of the CCDC146 mutation, we generated a mouse model ( Ccdc146 mut/mut ) with a similar mutation to that of the patient. Consistently, the Ccdc146 mut/mut mice exhibited infertility, characterized by significantly reduced sperm counts, diminished motility, and multiple defects in sperm heads and flagella. Furthermore, the levels of axonemal proteins, including DNAH17, DNAH1, and SPAG6, were significantly reduced in the sperm of Ccdc146 mut/mut mice. Additionally, both human and mouse CCDC146 interacted with intraflagellar transport protein 20 (IFT20), but this interaction was lost in the mutated versions, leading to the degradation of IFT20. This study identified a novel deleterious homozygous nonsense mutation in CCDC146 that causes male infertility, potentially by disrupting axonemal protein transportation. These findings offer valuable insights for genetic counseling and understanding the mechanisms underlying CCDC146 mutant-associated infertility in human males.


Subject(s)
Asthenozoospermia , Microtubule-Associated Proteins , Animals , Humans , Male , Mice , Asthenozoospermia/genetics , Codon, Nonsense , Homozygote , Infertility, Male/genetics , Mutation , Oligospermia/genetics , Sperm Motility/genetics , Spermatozoa , Microtubule-Associated Proteins/genetics
11.
Nat Cell Biol ; 26(9): 1520-1534, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39160291

ABSTRACT

Cells migrating through complex three-dimensional environments experience considerable physical challenges, including tensile stress and compression. To move, cells need to resist these forces while also squeezing the large nucleus through confined spaces. This requires highly coordinated cortical contractility. Microtubules can both resist compressive forces and sequester key actomyosin regulators to ensure appropriate activation of contractile forces. Yet, how these two roles are integrated to achieve nuclear transmigration in three dimensions is largely unknown. Here, we demonstrate that compression triggers reinforcement of a dedicated microtubule structure at the rear of the nucleus by the mechanoresponsive recruitment of cytoplasmic linker-associated proteins, which dynamically strengthens and repairs the lattice. These reinforced microtubules form the mechanostat: an adaptive feedback mechanism that allows the cell to both withstand compressive force and spatiotemporally organize contractility signalling pathways. The microtubule mechanostat facilitates nuclear positioning and coordinates force production to enable the cell to pass through constrictions. Disruption of the mechanostat imbalances cortical contractility, stalling migration and ultimately resulting in catastrophic cell rupture. Our findings reveal a role for microtubules as cellular sensors that detect and respond to compressive forces, enabling movement and ensuring survival in mechanically demanding environments.


Subject(s)
Cell Movement , Cell Nucleus , Microtubules , Microtubules/metabolism , Animals , Cell Nucleus/metabolism , Stress, Mechanical , Mechanotransduction, Cellular , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mice , Humans , Actomyosin/metabolism , Microfilament Proteins
12.
Curr Biol ; 34(18): 4170-4183.e9, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39181128

ABSTRACT

Proteome maintenance in contracting skeletal and cardiac muscles depends on the chaperone-regulating protein BAG3. Reduced BAG3 activity leads to muscle weakness and heart failure in animal models and patients. BAG3 and its chaperone partners recognize mechanically damaged muscle proteins and initiate their disposal through chaperone-assisted selective autophagy (CASA). However, molecular details of the force-dependent regulation of BAG3 have remained elusive so far. Here, we demonstrate that mechanical stress triggers the dephosphorylation of BAG3 in human muscle and in isolated cells. We identify force-regulated phospho-switches in BAG3 that control CASA complex assembly and CASA activity. Differential proteomics reveal RAB GTPases, which organize membrane traffic and fusion, as dephosphorylation-dependent interactors of BAG3. In fact, RAB7A and RAB11B are shown here to be essential for CASA in skeletal muscle cells. Moreover, BAG3 dephosphorylation is also observed upon induction of mitophagy, suggesting an involvement of the cochaperone in the RAB7A-dependent autophagic engulfment of damaged mitochondria in exercised muscle. Cooperation of BAG3 with RAB7A relies on a direct interaction of both proteins, which is regulated by the nucleotide state of the GTPase and by association with the autophagosome membrane protein LC3B. Finally, we provide evidence that BAG3 and RAB7A also cooperate in non-muscle cells and propose that overactivation of CASA in RAB7A-L129F patients contributes to the loss of peripheral neurons in Charcot-Marie-Tooth neuropathy.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Phosphorylation , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins/metabolism , Proteostasis , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Muscle, Skeletal/metabolism , Autophagy/physiology , Animals , Mice , Protein Transport
13.
Cell Death Dis ; 15(8): 591, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39143050

ABSTRACT

Neurons rely heavily on high mitochondrial metabolism to provide sufficient energy for proper development. However, it remains unclear how neurons maintain high oxidative phosphorylation (OXPHOS) during development. Mitophagy plays a pivotal role in maintaining mitochondrial quality and quantity. We herein describe that G protein-coupled receptor 50 (GPR50) is a novel mitophagy receptor, which harbors the LC3-interacting region (LIR) and is required in mitophagy under stress conditions. Although it does not localize in mitochondria under normal culturing conditions, GPR50 is recruited to the depolarized mitochondrial membrane upon mitophagy stress, which marks the mitochondrial portion and recruits the assembling autophagosomes, eventually facilitating the mitochondrial fragments to be engulfed by the autophagosomes. Mutations Δ502-505 and T532A attenuate GPR50-mediated mitophagy by disrupting the binding of GPR50 to LC3 and the mitochondrial recruitment of GPR50. Deficiency of GPR50 causes the accumulation of damaged mitochondria and disrupts OXPHOS, resulting in insufficient ATP production and excessive ROS generation, eventually impairing neuronal development. GPR50-deficient mice exhibit impaired social recognition, which is rescued by prenatal treatment with mitoQ, a mitochondrially antioxidant. The present study identifies GPR50 as a novel mitophagy receptor that is required to maintain mitochondrial OXPHOS in developing neurons.


Subject(s)
Mitochondria , Mitophagy , Neurons , Receptors, G-Protein-Coupled , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Neurons/metabolism , Mitochondria/metabolism , Mice , Humans , Oxidative Phosphorylation , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Reactive Oxygen Species/metabolism , Mice, Knockout , Neurogenesis
14.
Nat Commun ; 15(1): 7000, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143095

ABSTRACT

Mutations in the nuclear envelope (NE) protein lamin A/C (encoded by LMNA), cause a severe form of dilated cardiomyopathy (DCM) with early-onset life-threatening arrhythmias. However, molecular mechanisms underlying increased arrhythmogenesis in LMNA-related DCM (LMNA-DCM) remain largely unknown. Here we show that a frameshift mutation in LMNA causes abnormal Ca2+ handling, arrhythmias and disformed NE in LMNA-DCM patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). Mechanistically, lamin A interacts with sirtuin 1 (SIRT1) where mutant lamin A/C accelerates degradation of SIRT1, leading to mitochondrial dysfunction and oxidative stress. Elevated reactive oxygen species (ROS) then activates the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-ryanodine receptor 2 (RYR2) pathway and aggravates the accumulation of SUN1 in mutant iPSC-CMs, contributing to arrhythmias and NE deformation, respectively. Taken together, the lamin A/C deficiency-mediated ROS disorder is revealed as central to LMNA-DCM development. Manipulation of impaired SIRT1 activity and excessive oxidative stress is a potential future therapeutic strategy for LMNA-DCM.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Lamin Type A , Myocytes, Cardiac , Oxidative Stress , Reactive Oxygen Species , Sirtuin 1 , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Lamin Type A/metabolism , Lamin Type A/genetics , Induced Pluripotent Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Humans , Sirtuin 1/metabolism , Sirtuin 1/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Frameshift Mutation , Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Nuclear Envelope/metabolism , Mitochondria/metabolism , Male , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
15.
BMC Ophthalmol ; 24(1): 327, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107704

ABSTRACT

BACKGROUND: Occult Macular Dystrophy (OMD), primarily caused by retinitis pigmentosa 1-like 1 (RP1L1) variants, is a complex retinal disease characterised by progressive vision loss and a normal fundus appearance. This study aims to investigate the diverse phenotypic expressions and genotypic correlations of OMD in Chinese patients, including a rare case of Vitelliform Macular Dystrophy (VMD) associated with RP1L1. METHODS: We analysed seven OMD patients and one VMD patient, all with heterozygous pathogenic RP1L1 variants. Clinical assessments included Best Corrected Visual Acuity (BCVA), visual field testing, Spectral Domain Optical Coherence Tomography (SD-OCT), multifocal Electroretinograms (mfERGs), and microperimetry. Next-generation sequencing was utilised for genetic analysis. RESULTS: The OMD patients displayed a range of phenotypic variability. Most (5 out of 7) had the RP1L1 variant c.133 C > T; p.R45W, associated with central vision loss and specific patterns in SD-OCT and mfERG. Two patients exhibited different RP1L1 variants (c.3599G > T; p.G1200V and c.2880G > C; p.W960C), presenting milder phenotypes. SD-OCT revealed photoreceptor layer changes, with most patients showing decreased mfERG responses in the central rings. Interestingly, a unique case of VMD linked to the RP1L1 variant was observed, distinct from traditional OMD presentations. CONCLUSIONS: This study highlights the phenotypic diversity within OMD and the broader spectrum of RP1L1-associated macular dystrophies, including a novel association with VMD. The findings emphasise the complexity of RP1L1 variants in determining clinical manifestations, underscoring the need for comprehensive genetic and clinical evaluations in macular dystrophies.


Subject(s)
Electroretinography , Eye Proteins , Microtubule-Associated Proteins , Tomography, Optical Coherence , Visual Acuity , Vitelliform Macular Dystrophy , Humans , Male , Female , Tomography, Optical Coherence/methods , Adult , Middle Aged , Eye Proteins/genetics , Visual Acuity/physiology , Vitelliform Macular Dystrophy/genetics , Vitelliform Macular Dystrophy/physiopathology , Vitelliform Macular Dystrophy/diagnosis , Microtubule-Associated Proteins/genetics , Visual Fields/physiology , China/epidemiology , Young Adult , Visual Field Tests , Pedigree , Adolescent , Phenotype , Mutation , Macular Degeneration/genetics , Macular Degeneration/diagnosis , Macular Degeneration/physiopathology , Asian People/genetics , Aged , East Asian People
16.
Oral Oncol ; 157: 106983, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39126751

ABSTRACT

Sinonasal squamous cell carcinomas (SNSCCs) are uncommon and they are associated with adverse prognosis. HPV-associated SNSCCs and fusion-driven SNSCCs are particularly rare. A case of an HPV-associated SNSCC with a FGFR3::TACC3 fusion is thus presented; a brief review of the pertinent literature is also provided.


Subject(s)
Carcinoma, Squamous Cell , Receptor, Fibroblast Growth Factor, Type 3 , Humans , Receptor, Fibroblast Growth Factor, Type 3/genetics , Carcinoma, Squamous Cell/virology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Microtubule-Associated Proteins/genetics , Male , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Paranasal Sinus Neoplasms/virology , Paranasal Sinus Neoplasms/pathology , Paranasal Sinus Neoplasms/genetics , Middle Aged , Female
17.
Cells ; 13(16)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39195284

ABSTRACT

The AurkA serine/threonine kinase is a key regulator of cell division controlling mitotic entry, centrosome maturation, and chromosome segregation. The microtubule-associated protein TPX2 controls spindle assembly and is the main AurkA regulator, contributing to AurkA activation, localisation, and stabilisation. Since their identification, AurkA and TPX2 have been described as being overexpressed in cancer, with a significant correlation with highly proliferative and aneuploid tumours. Despite the frequent occurrence of AurkA/TPX2 co-overexpression in cancer, the investigation of their involvement in tumorigenesis and cancer therapy resistance mostly arises from studies focusing only on one at the time. Here, we review the existing literature and discuss the mitotic phenotypes described under conditions of AurkA, TPX2, or AurkA/TPX2 overexpression, to build a picture that may help clarify their oncogenic potential through the induction of chromosome instability. We highlight the relevance of the AurkA/TPX2 complex as an oncogenic unit, based on which we discuss recent strategies under development that aim at disrupting the complex as a promising therapeutic perspective.


Subject(s)
Aurora Kinase A , Microtubule-Associated Proteins , Neoplasms , Humans , Aurora Kinase A/metabolism , Aurora Kinase A/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Animals , Mitosis/genetics , Chromosome Aberrations , Chromosomal Instability/genetics , Gene Expression Regulation, Neoplastic
18.
Cell Rep ; 43(8): 114619, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39128005

ABSTRACT

Autophagosome formation initiated on the endoplasmic reticulum (ER)-associated omegasome requires LC3. Translational regulation of LC3 biosynthesis is unexplored. Here we demonstrate that LC3 mRNA is recruited to omegasomes by directly binding to the ER transmembrane Sigma-1 receptor (S1R). Cell-based and in vitro reconstitution experiments show that S1R interacts with the 3' UTR of LC3 mRNA and ribosomes to promote LC3 translation. Strikingly, the 3' UTR of LC3 is also required for LC3 protein lipidation, thereby linking the mRNA-3' UTR to LC3 function. An autophagy-defective S1R mutant responsible for amyotrophic lateral sclerosis cannot bind LC3 mRNA or induce LC3 translation. We propose a model wherein S1R de-represses LC3 mRNA via its 3' UTR at the ER, enabling LC3 biosynthesis and lipidation. Because several other LC3-related proteins use the same mechanism, our data reveal a conserved pathway for localized translation essential for autophagosome biogenesis with insights illuminating the molecular basis of a neurodegenerative disease.


Subject(s)
3' Untranslated Regions , Autophagy , Endoplasmic Reticulum , Microtubule-Associated Proteins , Protein Biosynthesis , RNA, Messenger , Receptors, sigma , Sigma-1 Receptor , Receptors, sigma/metabolism , Receptors, sigma/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Endoplasmic Reticulum/metabolism , Humans , RNA, Messenger/metabolism , RNA, Messenger/genetics , 3' Untranslated Regions/genetics , Ribosomes/metabolism , Animals , Autophagosomes/metabolism , HeLa Cells
19.
Curr Biol ; 34(15): R724-R726, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39106827

ABSTRACT

The hormone leptin is critical for regulation of food intake, energy expenditure and overall metabolism. However, the mechanisms that promote leptin secretion from adipocytes in response to nutrient surplus and limit its secretion during nutrient scarcity are unclear. New work reveals that the autophagy protein Atg8/LC3 has a bidirectional role in leptin secretion, both facilitating and limiting its release.


Subject(s)
Autophagy , Leptin , Autophagy/physiology , Animals , Leptin/metabolism , Nutrients/metabolism , Adipocytes/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Energy Metabolism , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics
20.
Nat Commun ; 15(1): 6509, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095354

ABSTRACT

Microtubule organization in cells relies on targeting mechanisms. Cytoplasmic linker proteins (CLIPs) and CLIP-associated proteins (CLASPs) are key regulators of microtubule organization, yet the underlying mechanisms remain elusive. Here, we reveal that the C-terminal domain of CLASP2 interacts with a common motif found in several CLASP-binding proteins. This interaction drives the dynamic localization of CLASP2 to distinct cellular compartments, where CLASP2 accumulates in protein condensates at the cell cortex or the microtubule plus end. These condensates physically contact each other via CLASP2-mediated competitive binding, determining cortical microtubule targeting. The phosphorylation of CLASP2 modulates the dynamics of the condensate-condensate interaction and spatiotemporally navigates microtubule growth. Moreover, we identify additional CLASP-interacting proteins that are involved in condensate contacts in a CLASP2-dependent manner, uncovering a general mechanism governing microtubule targeting. Our findings not only unveil a tunable multiphase system regulating microtubule organization, but also offer general mechanistic insights into intricate protein-protein interactions at the mesoscale level.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Protein Binding , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Humans , Phosphorylation , Binding, Competitive , HeLa Cells , Biomolecular Condensates/metabolism , HEK293 Cells , Animals
SELECTION OF CITATIONS
SEARCH DETAIL