Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(4): 107124, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38432637

ABSTRACT

Rab35 (Ras-associated binding protein) is a small GTPase that regulates endosomal membrane trafficking and functions in cell polarity, cytokinesis, and growth factor signaling. Altered Rab35 function contributes to progression of glioblastoma, defects in primary cilia formation, and altered cytokinesis. Here, we report a pediatric patient with global developmental delay, hydrocephalus, a Dandy-Walker malformation, axial hypotonia with peripheral hypertonia, visual problems, and conductive hearing impairment. Exome sequencing identified a homozygous missense variant in the GTPase fold of RAB35 (c.80G>A; p.R27H) as the most likely candidate. Functional analysis of the R27H-Rab35 variant protein revealed enhanced interaction with its guanine-nucleotide exchange factor, DENND1A and decreased interaction with a known effector, MICAL1, indicating that the protein is in an inactive conformation. Cellular expression of the variant drives the activation of Arf6, a small GTPase under negative regulatory control of Rab35. Importantly, variant expression leads to delayed cytokinesis and altered length, number, and Arl13b composition of primary cilia, known factors in neurodevelopmental disease. Our findings provide evidence of altered Rab35 function as a causative factor of a neurodevelopmental disorder.


Subject(s)
Mutation, Missense , Neurodevelopmental Disorders , rab GTP-Binding Proteins , Female , Humans , Male , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Cell Line , Cilia/metabolism , Cilia/genetics , Cilia/pathology , Cytokinesis/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Loss of Function Mutation , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Pedigree , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Models, Molecular , Protein Structure, Tertiary
3.
J Biol Chem ; 298(9): 102320, 2022 09.
Article in English | MEDLINE | ID: mdl-35933016

ABSTRACT

Autosomal recessive spastic ataxia of Charlevoix-Saguenay is a fatal brain disorder featuring cerebellar neurodegeneration leading to spasticity and ataxia. This disease is caused by mutations in the SACS gene that encodes sacsin, a massive 4579-amino acid protein with multiple modular domains. However, molecular details of the function of sacsin are not clear. Here, using live cell imaging and biochemistry, we demonstrate that sacsin binds to microtubules and regulates microtubule dynamics. Loss of sacsin function in various cell types, including knockdown and KO primary neurons and patient fibroblasts, leads to alterations in lysosomal transport, positioning, function, and reformation following autophagy. Each of these phenotypic changes is consistent with altered microtubule dynamics. We further show the effects of sacsin are mediated at least in part through interactions with JIP3, an adapter for microtubule motors. These data reveal a new function for sacsin that explains its previously reported roles and phenotypes.


Subject(s)
Heat-Shock Proteins , Lysosomes , Microtubules , Muscle Spasticity , Spinocerebellar Ataxias , Amino Acid Sequence , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Lysosomes/genetics , Lysosomes/metabolism , Microtubules/genetics , Microtubules/metabolism , Muscle Spasticity/genetics , Muscle Spasticity/metabolism , Mutation , Spinocerebellar Ataxias/congenital , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
4.
J Biol Chem ; 296: 100306, 2021.
Article in English | MEDLINE | ID: mdl-33476648

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of COVID-19, so understanding its biology and infection mechanisms is critical to facing this major medical challenge. SARS-CoV-2 is known to use its spike glycoprotein to interact with the cell surface as a first step in the infection process. As for other coronaviruses, it is likely that SARS-CoV-2 next undergoes endocytosis, but whether or not this is required for infectivity and the precise endocytic mechanism used are unknown. Using purified spike glycoprotein and lentivirus pseudotyped with spike glycoprotein, a common model of SARS-CoV-2 infectivity, we now demonstrate that after engagement with the plasma membrane, SARS-CoV-2 undergoes rapid, clathrin-mediated endocytosis. This suggests that transfer of viral RNA to the cell cytosol occurs from the lumen of the endosomal system. Importantly, we further demonstrate that knockdown of clathrin heavy chain, which blocks clathrin-mediated endocytosis, reduces viral infectivity. These discoveries reveal that SARS-CoV-2 uses clathrin-mediated endocytosis to gain access into cells and suggests that this process is a key aspect of virus infectivity.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Clathrin Heavy Chains/genetics , Endocytosis/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Virus Internalization/drug effects , A549 Cells , Angiotensin-Converting Enzyme 2/metabolism , Animals , Chlorocebus aethiops , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Endosomes/virology , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Hydrazones/pharmacology , Lentivirus/genetics , Lentivirus/metabolism , Protein Binding/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Signal Transduction , Spike Glycoprotein, Coronavirus/metabolism , Sulfonamides/pharmacology , Thiazolidines/pharmacology , Vero Cells
5.
Br J Psychiatry ; 216(6): 331-337, 2020 06.
Article in English | MEDLINE | ID: mdl-31046864

ABSTRACT

BACKGROUND: Around 60 000 people in England live in mental health supported accommodation. There are three main types: residential care, supported housing and floating outreach. Supported housing and floating outreach aim to support service users in moving on to more independent accommodation within 2 years, but there has been little research investigating their effectiveness. AIMS: A 30-month prospective cohort study investigating outcomes for users of mental health supported accommodation. METHOD: We used random sampling, accounting for relevant geographical variation factors, to recruit 87 services (22 residential care, 35 supported housing and 30 floating outreach) and 619 service users (residential care 159, supported housing 251, floating outreach 209) across England. We contacted services every 3 months to investigate the proportion of service users who successfully moved on to more independent accommodation. Multilevel modelling was used to estimate how much of the outcome and cost variations were due to service type and quality, after accounting for service-user characteristics. RESULTS: Overall 243/586 participants successfully moved on (residential care 15/146, supported housing 96/244, floating outreach 132/196). This was most likely for floating outreach service users (versus residential care: odds ratio 7.96, 95% CI 2.92-21.69, P < 0.001; versus supported housing: odds ratio 2.74, 95% CI 1.01-7.41, P < 0.001) and was associated with reduced costs of care and two aspects of service quality: promotion of human rights and recovery-based practice. CONCLUSIONS: Most people do not move on from supported accommodation within the expected time frame. Greater focus on human rights and recovery-based practice may increase service effectiveness.


Subject(s)
Housing/statistics & numerical data , Mental Disorders , Mental Health Services/statistics & numerical data , England , Female , Health Care Costs , Housing/economics , Human Rights , Humans , Male , Mental Disorders/economics , Mental Health Services/economics , Odds Ratio , Prospective Studies , Time Factors
6.
J Biol Chem ; 293(12): 4566-4574, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29352104

ABSTRACT

Rab GTPases are key regulators of membrane trafficking, and many are activated by guanine nucleotide exchange factors bearing a differentially expressed in normal and neoplastic cells (DENN) domain. By activating the small GTPase Rab12, DENN domain-containing protein 3 (DENND3) functions in autophagy. Here, we identified a structural domain (which we name PHenn) containing a pleckstrin homology subdomain that binds actin and is required for DENND3 function in autophagy. We found that a hydrophobic patch on an extended ß-turn of the PHenn domain mediates an intramolecular interaction with the DENN domain of DENND3. We also show that DENND3 binds actin through a surface of positively charged residues on the PHenn domain. Substitutions that blocked either DENN or actin binding compromised the role of DENND3 in autophagy. These results provide new mechanistic insight into the structural determinants regulating DENND3 in autophagy and lay the foundation for future investigations of the DENN protein family.


Subject(s)
Actins/metabolism , Autophagy , Blood Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phosphoproteins/metabolism , rab GTP-Binding Proteins/metabolism , Actins/chemistry , Actins/genetics , Blood Proteins/chemistry , Crystallography, X-Ray , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Phosphoproteins/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics
7.
J Biol Chem ; 293(33): 12832-12842, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29945973

ABSTRACT

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a neurodegenerative disease that is caused by mutations in the SACS gene. The product of this gene is a very large 520-kDa cytoplasmic protein, sacsin, with a ubiquitin-like (Ubl) domain at the N terminus followed by three large sacsin internal repeat (SIRPT) supradomains and C-terminal J and HEPN domains. The SIRPTs are predicted to contain Hsp90-like domains, suggesting a potential chaperone activity. In this work, we report the structures of the Hsp90-like Sr1 domain of SIRPT1 and the N-terminal Ubl domain determined at 1.55- and 2.1-Å resolutions, respectively. The Ubl domain crystallized as a swapped dimer that could be relevant in the context of full-length protein. The Sr1 domain displays the Bergerat protein fold with a characteristic nucleotide-binding pocket, although it binds nucleotides with very low affinity. The Sr1 structure reveals that ARSACS-causing missense mutations (R272H, R272C, and T201K) disrupt protein folding, most likely leading to sacsin degradation. This work lends structural support to the view of sacsin as a molecular chaperone and provides a framework for future studies of this protein.


Subject(s)
Heat-Shock Proteins/chemistry , Mutation, Missense , Protein Folding , Amino Acid Substitution , Crystallography, X-Ray , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Muscle Spasticity/genetics , Muscle Spasticity/metabolism , Protein Domains , Spinocerebellar Ataxias/congenital , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
8.
Am J Hum Genet ; 99(6): 1359-1367, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27866705

ABSTRACT

Epileptic encephalopathies are a catastrophic group of epilepsies characterized by refractory seizures and cognitive arrest, often resulting from abnormal brain development. Here, we have identified an epileptic encephalopathy additionally featuring cerebral calcifications and coarse facial features caused by recessive loss-of-function mutations in DENND5A. DENND5A contains a DENN domain, an evolutionarily ancient enzymatic module conferring guanine nucleotide exchange factor (GEF) activity to multiple proteins serving as GEFs for Rabs, which are key regulators of membrane trafficking. DENND5A is detected predominantly in neuronal tissues, and its highest levels occur during development. Knockdown of DENND5A leads to striking alterations in neuronal development. Mechanistically, these changes appear to result from upregulation of neurotrophin receptors, leading to enhanced downstream signaling. Thus, we have identified a link between a DENN domain protein and neuronal development, dysfunction of which is responsible for a form of epileptic encephalopathy.


Subject(s)
Brain/pathology , Epilepsy/genetics , Mutation , rab GTP-Binding Proteins/genetics , Adolescent , Animals , Child , Consanguinity , Female , Guanine Nucleotide Exchange Factors , Humans , Male , Neurons/metabolism , PC12 Cells , Pedigree , Rats
9.
EMBO Rep ; 18(12): 2119-2130, 2017 12.
Article in English | MEDLINE | ID: mdl-29030480

ABSTRACT

Epidermal growth factor (EGF) activates the EGF receptor (EGFR) and stimulates its internalization and trafficking to lysosomes for degradation. However, a percentage of EGFR undergoes ligand-independent endocytosis and is rapidly recycled back to the plasma membrane. Importantly, alterations in EGFR recycling are a common hallmark of cancer, and yet, our understanding of the machineries controlling the fate of endocytosed EGFR is incomplete. Intersectin-s is a multi-domain adaptor protein that is required for internalization of EGFR Here, we discover that intersectin-s binds DENND2B, a guanine nucleotide exchange factor for the exocytic GTPase Rab13, and this interaction promotes recycling of ligand-free EGFR to the cell surface. Intriguingly, upon EGF treatment, DENND2B is phosphorylated by protein kinase D and dissociates from intersectin-s, allowing for receptor targeting to degradation. Our study thus reveals a novel mechanism controlling the fate of internalized EGFR with important implications for cancer.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Cell Membrane/metabolism , Endocytosis , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/genetics , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Neoplasms/physiopathology , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Protein Transport , Tumor Suppressor Proteins/genetics , rab GTP-Binding Proteins/metabolism
10.
J Biol Chem ; 292(17): 7274-7282, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28249939

ABSTRACT

The Rab family of small GTPases functions in multiple aspects of cellular membrane trafficking. Proteins bearing a differentially expressed in normal and neoplastic cells (DENN) domain have emerged as the largest family of Rab-activating guanine nucleotide exchange factors (GEFs). Rab12 functions in the initiation of starvation-induced autophagy, and our previous work revealed that its activator, DENN domain-containing protein 3 (DENND3), is phosphorylated and activated upon starvation. However, how the GEF activity of DENND3 toward Rab12 is regulated at the molecular level is still not understood. Here, we combine size-exclusion chromatography, Förster resonance energy transfer, pulldown, and in vitro GEF assays to demonstrate that regulation of GEF activity is achieved through an intramolecular interaction that is controlled by a key residue in DENND3, tyrosine 940. Our study sheds light on the regulation of Rab12 activation and lays the groundwork for characterizing the regulation of other DENN domain-containing proteins.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Autophagy , Cell Membrane/metabolism , Chromatography , Fluorescence Resonance Energy Transfer , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Mice , Microscopy, Fluorescence , Mutation , Phosphorylation , Protein Binding , Protein Domains , Tyrosine/chemistry
12.
BMC Psychiatry ; 18(1): 128, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29764420

ABSTRACT

BACKGROUND: Post-deinstitutionalisation, mental health supported accommodation services have been implemented widely. The available research evidence is heterogeneous in nature and resistant to synthesis attempts, leaving researchers and policy makers with no clear summary what works and for whom. In this context, we undertook a comprehensive systematic review of quantitative studies in order to synthesise the current evidence on mental health and psychosocial outcomes for individuals residing in mental health supported accommodation services. METHODS: Using a combination of electronic database searches, hand searches, forward-backward snowballing and article recommendations from an expert panel, 115 papers were identified for review. Data extraction and quality assessments were conducted, and 33 articles were excluded due to low quality, leaving 82 papers in the final review. Variation in terminology and service characteristics made the comparison of service models unfeasible. As such, findings were presented according to the following sub-groups: 'Homeless', 'Deinstitutionalisation' and 'General Severe Mental Illness (SMI)'. RESULTS: Results were mixed, reflecting the heterogeneity of the supported accommodation literature, in terms of research quality, experimental design, population, service types and outcomes assessed. There is some evidence that supported accommodation is effective across a range of psychosocial outcomes. The most robust evidence supports the effectiveness of the permanent supported accommodation model for homeless SMI in generating improvements in housing retention and stability, and appropriate use of clinical services over time, and for other forms of supported accommodation for deinstitutionalised populations in reducing hospitalisation rates and improving appropriate service use. The evidence base for general SMI populations is less developed, and requires further research. CONCLUSIONS: A lack of high-quality experimental studies, definitional inconsistency and poor reporting continue to stymie our ability to identify effective supported accommodation models and practices. The authors recommend improved reporting standards and the prioritisation of experimental studies that compare outcomes across different service models.


Subject(s)
Deinstitutionalization/methods , Housing , Mental Disorders/rehabilitation , Mental Health Services , Psychiatric Rehabilitation/methods , Humans , Psychiatric Rehabilitation/psychology
13.
Mol Cell ; 37(3): 370-82, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20159556

ABSTRACT

The DENN domain is an evolutionarily ancient protein module. Mutations in the DENN domain cause developmental defects in plants and human diseases, yet the function of this common module is unknown. We now demonstrate that the connecdenn/DENND1A DENN domain functions as a guanine nucleotide exchange factor (GEF) for Rab35 to regulate endosomal membrane trafficking. Loss of Rab35 activity causes an enlargement of early endosomes and inhibits MHC class I recycling. Moreover, it prevents early endosomal recruitment of EHD1, a common component of tubules involved in endosomal cargo recycling. Our data reveal an enzymatic activity for a DENN domain and demonstrate that distinct Rab GTPases can recruit a common protein machinery to various sites within the endosomal network to establish cargo-selective recycling pathways.


Subject(s)
Endosomes/metabolism , Guanine Nucleotide Exchange Factors/physiology , rab GTP-Binding Proteins/physiology , Animals , Biological Transport , COS Cells , Chlorocebus aethiops , Clathrin-Coated Vesicles/metabolism , Endocytosis , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Humans , Protein Interaction Mapping , Protein Structure, Tertiary , Rats , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/metabolism
14.
J Biol Chem ; 291(19): 9929-37, 2016 May 06.
Article in English | MEDLINE | ID: mdl-27044746

ABSTRACT

The members of the Rab family of GTPases are master regulators of cellular membrane trafficking. With ∼70 members in humans, Rabs have been implicated in all steps of membrane trafficking ranging from vesicle formation and transport to vesicle docking/tethering and fusion. Vesicle trafficking controls the localization and levels of a myriad of proteins, thus regulating cellular functions including proliferation, metabolism, cell-cell adhesion, and cell migration. It is therefore not surprising that impairment of Rab pathways is associated with diseases including cancer. In this review, we highlight evidence supporting the role of Rab13 as a potent driver of cancer progression.


Subject(s)
Cell Membrane/metabolism , Cell Proliferation , Neoplasm Proteins/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism , Animals , Biological Transport, Active , Cell Membrane/genetics , Cell Membrane/pathology , Humans , Neoplasm Proteins/genetics , Neoplasms , rab GTP-Binding Proteins/genetics
15.
J Biol Chem ; 291(20): 10726-35, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26969162

ABSTRACT

Rab GTPases are critical regulators of membrane trafficking. The canonical view is that Rabs are soluble in their inactive GDP-bound form, and only upon activation and conversion to their GTP-bound state are they anchored to membranes through membrane insertion of a C-terminal prenyl group. Here we demonstrate that C-terminal prenylation is not required for Rab13 to associate with and traffic on vesicles. Instead, inactive Rab13 appears to associate with vesicles via protein-protein interactions. Only following activation does Rab13 associate with the plasma membrane, presumably with insertion of the C-terminal prenyl group into the membrane.


Subject(s)
rab GTP-Binding Proteins/metabolism , Amino Acid Substitution , Animals , Cell Line , Cell Membrane Structures/metabolism , Cytoplasmic Vesicles/metabolism , Endosomes/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , HEK293 Cells , Humans , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Prenylation , Protein Transport , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Cell Analysis , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics
16.
Hum Mol Genet ; 24(3): 727-39, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25260547

ABSTRACT

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS [MIM 270550]) is an early-onset neurodegenerative disorder caused by mutations in the SACS gene. Over 170 SACS mutations have been reported worldwide and are thought to cause loss of function of sacsin, a poorly characterized and massive 520 kDa protein. To establish an animal model and to examine the pathophysiological basis of ARSACS, we generated Sacs knockout (Sacs(-/-)) mice. Null animals displayed an abnormal gait with progressive motor, cerebellar and peripheral nerve dysfunctions highly reminiscent of ARSACS. These clinical features were accompanied by an early onset, progressive loss of cerebellar Purkinje cells followed by spinal motor neuron loss and peripheral neuropathy. Importantly, loss of sacsin function resulted in abnormal accumulation of non-phosphorylated neurofilament (NF) bundles in the somatodendritic regions of vulnerable neuronal populations, a phenotype also observed in an ARSACS brain. Moreover, motor neurons cultured from Sacs(-/-) embryos exhibited a similar NF rearrangement with significant reduction in mitochondrial motility and elongated mitochondria. The data points to alterations in the NF cytoskeleton and defects in mitochondrial dynamics as the underlying pathophysiological basis of ARSACS.


Subject(s)
Heat-Shock Proteins/genetics , Mitochondria/pathology , Motor Neurons/pathology , Muscle Spasticity/physiopathology , Purkinje Cells/pathology , Spinocerebellar Ataxias/congenital , Animals , Disease Models, Animal , Heat-Shock Proteins/metabolism , Humans , Intermediate Filaments/pathology , Mice , Mice, Knockout , Motor Neurons/cytology , Muscle Spasticity/genetics , Purkinje Cells/metabolism , Pyramidal Tracts/pathology , Spine/pathology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Tissue Culture Techniques
17.
J Cell Sci ; 128(12): 2229-35, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25964652

ABSTRACT

Synaptotagmin 1 (Syt1) is a synaptic vesicle protein that is important for the kinetics of both exocytosis and endocytosis, and is thus a candidate molecule to link these two processes. Although the tandem Ca(2+)-binding C2 domains of Syt1 have important roles in exocytosis and endocytosis, the function of the conserved juxtamembrane (jxm) linker region has yet to be determined. We now demonstrate that the jxm region of Syt1 interacts directly with the pleckstrin homology (PH) domain of the endocytic protein dynamin 1. By using cell-attached capacitance recordings with millisecond time resolution to monitor clathrin-mediated endocytosis of single vesicles in neuroendocrine chromaffin cells, we find that loss of this interaction prolongs the lifetime of the fission pore leading to defects in the dynamics of vesicle fission. These results indicate a previously undescribed interaction between two major regulatory proteins in the secretory vesicle cycle and that this interaction regulates endocytosis.


Subject(s)
Brain/metabolism , Chromaffin Cells/metabolism , Dynamin I/metabolism , Synaptic Vesicles/physiology , Synaptotagmin I/physiology , Amino Acid Sequence , Animals , Blotting, Western , Brain/cytology , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Chromaffin Cells/cytology , Clathrin/metabolism , Endocytosis/physiology , Exocytosis/physiology , Female , Humans , Immunoprecipitation , Male , Mice , Mice, Knockout , Molecular Sequence Data , Protein Interaction Domains and Motifs , Rats , Sequence Homology, Amino Acid , Synapses/physiology
18.
EMBO Rep ; 16(6): 709-18, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25925668

ABSTRACT

Unc-51-like kinases (ULKs) are the most upstream kinases in the initiation of autophagy, yet the molecular mechanisms underlying their function are poorly understood. We report a new role for ULK in the induction of autophagy. ULK-mediated phosphorylation of the guanine nucleotide exchange factor DENND3 at serines 554 and 572 upregulates its GEF activity toward the small GTPase Rab12. Through binding to LC3 and associating with LC3-positive autophagosomes, active Rab12 facilitates autophagosome trafficking, thus establishing a crucial role for the ULK/DENND3/Rab12 axis in starvation-induced autophagy.


Subject(s)
Autophagy , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , rab GTP-Binding Proteins/metabolism , Autophagy-Related Protein-1 Homolog , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phagosomes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Transport , RNA, Small Interfering , rab GTP-Binding Proteins/genetics
19.
EMBO Rep ; 16(1): 79-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25427558

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of dominant-inherited Parkinson's disease (PD), and yet we do not fully understand the physiological function(s) of LRRK2. Various components of the clathrin machinery have been recently found mutated in familial forms of PD. Here, we provide molecular insight into the association of LRRK2 with the clathrin machinery. We report that through its GTPase domain, LRRK2 binds directly to clathrin-light chains (CLCs). Using genome-edited HA-LRRK2 cells, we localize LRRK2 to endosomes on the degradative pathway, where it partially co-localizes with CLCs. Knockdown of CLCs and/or LRRK2 enhances the activation of the small GTPase Rac1, leading to alterations in cell morphology, including the disruption of neuronal dendritic spines. In Drosphila, a minimal rough eye phenotype caused by overexpression of Rac1, is dramatically enhanced by loss of function of CLC and LRRK2 homologues, confirming the importance of this pathway in vivo. Our data identify a new pathway in which CLCs function with LRRK2 to control Rac1 activation on endosomes, providing a new link between the clathrin machinery, the cytoskeleton and PD.


Subject(s)
Clathrin Light Chains/metabolism , Endosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Brain/cytology , Brain/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Clathrin Light Chains/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Eye/metabolism , Eye/pathology , Gene Knockdown Techniques , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Molecular Sequence Data , Neurons/metabolism , Neurons/pathology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Rats , rac1 GTP-Binding Protein/genetics
20.
Mol Cell ; 36(6): 1034-47, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-20064468

ABSTRACT

Mutations in the parkin gene are responsible for a common inherited form of Parkinson's disease (PD). Parkin is a RING-type E3 ubiquitin ligase with an N-terminal ubiquitin-like domain (Ubl). We report here that the parkin Ubl binds SH3 domains from endocytic BAR proteins such as endophilin-A with an affinity comparable to proline-rich domains (PRDs) from well-established SH3 partners. The NMR structure of the Ubl-SH3 complex identifies the PaRK extension, a unique C-terminal motif in the parkin Ubl required for SH3 binding and for parkin-mediated ubiquitination of endophilin-A in vitro. In nerve terminals, conditions that promote phosphorylation enhance the interaction between parkin and endophilin-A and increase the levels of ubiquitinated proteins within PRD-associated synaptic protein complexes in wild-type but not parkin knockout brain. The findings identify a pathway for the recruitment of synaptic substrates to parkin with the potential to explain the defects in synaptic transmission observed in recessive forms of PD.


Subject(s)
Acyltransferases/metabolism , Protein Isoforms/metabolism , Synapses/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , src Homology Domains , Acyltransferases/genetics , Amino Acid Sequence , Animals , Cell Line , Glutamic Acid/metabolism , Humans , Mice , Models, Molecular , Molecular Sequence Data , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Protein Binding , Protein Isoforms/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Synaptic Transmission/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL