Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 44(6): 226-244, 2024.
Article in English | MEDLINE | ID: mdl-38828998

ABSTRACT

TIMM50 is a core subunit of the TIM23 complex, the mitochondrial inner membrane translocase responsible for the import of pre-sequence-containing precursors into the mitochondrial matrix and inner membrane. Here we describe a mitochondrial disease patient who is homozygous for a novel variant in TIMM50 and establish the first proteomic map of mitochondrial disease associated with TIMM50 dysfunction. We demonstrate that TIMM50 pathogenic variants reduce the levels and activity of endogenous TIM23 complex, which significantly impacts the mitochondrial proteome, resulting in a combined oxidative phosphorylation (OXPHOS) defect and changes to mitochondrial ultrastructure. Using proteomic data sets from TIMM50 patient fibroblasts and a TIMM50 HEK293 cell model of disease, we reveal that laterally released substrates imported via the TIM23SORT complex pathway are most sensitive to loss of TIMM50. Proteins involved in OXPHOS and mitochondrial ultrastructure are enriched in the TIM23SORT substrate pool, providing a biochemical mechanism for the specific defects in TIMM50-associated mitochondrial disease patients. These results highlight the power of using proteomics to elucidate molecular mechanisms of disease and uncovering novel features of fundamental biology, with the implication that human TIMM50 may have a more pronounced role in lateral insertion than previously understood.


Subject(s)
Mitochondria , Mitochondrial Diseases , Mitochondrial Precursor Protein Import Complex Proteins , Oxidative Phosphorylation , Protein Transport , Humans , Mitochondrial Precursor Protein Import Complex Proteins/metabolism , HEK293 Cells , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Diseases/genetics , Proteomics/methods , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Fibroblasts/metabolism , Mitochondrial Membranes/metabolism , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mutation/genetics
2.
Clin Genet ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38779778

ABSTRACT

Premature ovarian insufficiency is a common form of female infertility affecting up to 4% of women and characterised by amenorrhea with elevated gonadotropin before the age of 40. Oocytes require controlled DNA breakage and repair for homologous recombination and the maintenance of oocyte integrity. Biallelic disruption of the DNA damage repair gene, Fanconi anemia complementation group A (FANCA), is a common cause of Fanconi anaemia, a syndrome characterised by bone marrow failure, cancer predisposition, physical anomalies and POI. There is ongoing dispute about the role of heterozygous FANCA variants in POI pathogenesis, with insufficient evidence supporting causation. Here, we have identified biallelic FANCA variants in French sisters presenting with POI, including a novel missense variant of uncertain significance and a likely pathogenic deletion that initially evaded detection. Functional studies indicated no discernible effect on DNA damage sensitivity in patient lymphoblasts. These novel FANCA variants add evidence that heterozygous loss of one allele is insufficient to cause DNA damage sensitivity and POI. We propose that intragenic deletions, that are relatively common in FANCA, may be missed without careful analysis, and could explain the presumed causation of heterozygous variants. Accurate variant curation is critical to optimise patient care and outcomes.

3.
J Cell Biol ; 223(3)2024 03 04.
Article in English | MEDLINE | ID: mdl-38270563

ABSTRACT

CLPB is a mitochondrial intermembrane space AAA+ domain-containing disaggregase. CLPB mutations are associated with 3-methylglutaconic aciduria and neutropenia; however, the molecular mechanism underscoring disease and the contribution of CLPB substrates to disease pathology remains unknown. Interactions between CLPB and mitochondrial quality control (QC) factors, including PARL and OPA1, have been reported, hinting at dysregulation of organelle QC in disease. Utilizing proteomic and biochemical approaches, we show a stress-specific aggregation phenotype in a CLPB-null environment and define the CLPB substrate profile. We illustrate an interplay between intermembrane space proteins including CLPB, HAX1, HTRA2, and the inner membrane quality control proteins (STOML2, PARL, YME1L1; SPY complex), with CLPB deficiency impeding SPY complex function by virtue of protein aggregation in the intermembrane space. We conclude that there is an interdependency of mitochondrial QC components at the intermembrane space/inner membrane interface, and perturbations to this network may underscore CLPB disease pathology.


Subject(s)
Endopeptidase Clp , Intracellular Membranes , Membrane Proteins , Membrane Proteins/genetics , Mitochondria/genetics , Proteolysis , Proteomics , Humans , Endopeptidase Clp/genetics
4.
EMBO Rep ; 24(8): e56430, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37272231

ABSTRACT

Human Tim8a and Tim8b are paralogous intermembrane space proteins of the small TIM chaperone family. Yeast small TIMs function in the trafficking of proteins to the outer and inner mitochondrial membranes. This putative import function for hTim8a and hTim8b has been challenged in human models, but their precise molecular function(s) remains undefined. Likewise, the necessity for human cells to encode two Tim8 proteins and whether any potential redundancy exists is unclear. We demonstrate that hTim8a and hTim8b function in the assembly of cytochrome c oxidase (Complex IV). Using affinity enrichment mass spectrometry, we define the interaction network of hTim8a, hTim8b and hTim13, identifying subunits and assembly factors of the Complex IV COX2 module. hTim8-deficient cells have a COX2 and COX3 module defect and exhibit an accumulation of the Complex IV S2 subcomplex. These data suggest that hTim8a and hTim8b function in assembly of Complex IV via interactions with intermediate-assembly subcomplexes. We propose that hTim8-hTim13 complexes are auxiliary assembly factors involved in the formation of the Complex IV S3 subcomplex during assembly of mature Complex IV.


Subject(s)
Mitochondrial Membrane Transport Proteins , Saccharomyces cerevisiae Proteins , Humans , Mitochondrial Membrane Transport Proteins/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Cyclooxygenase 2/analysis , Cyclooxygenase 2/metabolism , Mitochondrial Membranes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Mitochondrial Proteins/metabolism
5.
Cell ; 186(10): 2219-2237.e29, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172566

ABSTRACT

The Commander complex is required for endosomal recycling of diverse transmembrane cargos and is mutated in Ritscher-Schinzel syndrome. It comprises two sub-assemblies: Retriever composed of VPS35L, VPS26C, and VPS29; and the CCC complex which contains twelve subunits: COMMD1-COMMD10 and the coiled-coil domain-containing (CCDC) proteins CCDC22 and CCDC93. Combining X-ray crystallography, electron cryomicroscopy, and in silico predictions, we have assembled a complete structural model of Commander. Retriever is distantly related to the endosomal Retromer complex but has unique features preventing the shared VPS29 subunit from interacting with Retromer-associated factors. The COMMD proteins form a distinctive hetero-decameric ring stabilized by extensive interactions with CCDC22 and CCDC93. These adopt a coiled-coil structure that connects the CCC and Retriever assemblies and recruits a 16th subunit, DENND10, to form the complete Commander complex. The structure allows mapping of disease-causing mutations and reveals the molecular features required for the function of this evolutionarily conserved trafficking machinery.


Subject(s)
Abnormalities, Multiple , Craniofacial Abnormalities , Multiprotein Complexes , Humans , Endosomes/metabolism , Protein Transport , Proteins/metabolism , Multiprotein Complexes/metabolism
6.
Front Cell Infect Microbiol ; 12: 894644, 2022.
Article in English | MEDLINE | ID: mdl-35646729

ABSTRACT

Streptococcus pneumoniae (Spn) is the primary agent of community-acquired pneumonia. Neutrophils are innate immune cells that are essential for bacterial clearance during pneumococcal pneumonia but can also do harm to host tissue. Neutrophil migration in pneumococcal pneumonia is therefore a major determinant of host disease outcomes. During Spn infection, detection of the bacterium leads to an increase in proinflammatory signals and subsequent expression of integrins and ligands on both the neutrophil as well as endothelial and epithelial cells. These integrins and ligands mediate the tethering and migration of the neutrophil from the bloodstream to the site of infection. A gradient of host-derived and bacterial-derived chemoattractants contribute to targeted movement of neutrophils. During pneumococcal pneumonia, neutrophils are rapidly recruited to the pulmonary space, but studies show that some of the canonical neutrophil migratory machinery is dispensable. Investigation of neutrophil migration is necessary for us to understand the dynamics of pneumococcal infection. Here, we summarize what is known about the pathways that lead to migration of the neutrophil from the capillaries to the lung during pneumococcal infection.


Subject(s)
Pneumonia, Pneumococcal , Humans , Integrins , Ligands , Neutrophil Infiltration , Streptococcus pneumoniae
7.
Mol Microbiol ; 117(5): 1245-1262, 2022 05.
Article in English | MEDLINE | ID: mdl-35403274

ABSTRACT

Infection with Plasmodium falciparum parasites results in approximately 627,000 deaths from malaria annually. Key to the parasite's success is their ability to invade and subsequently grow within human erythrocytes. Parasite proteins involved in parasite invasion and proliferation are therefore intrinsically of great interest, as targeting these proteins could provide novel means of therapeutic intervention. One such protein is P113 which has been reported to be both an invasion protein and an intracellular protein located within the parasitophorous vacuole (PV). The PV is delimited by a membrane (PVM) across which a plethora of parasite-specific proteins are exported via the Plasmodium Translocon of Exported proteins (PTEX) into the erythrocyte to enact various immune evasion functions. To better understand the role of P113 we isolated its binding partners from in vitro cultures of P. falciparum. We detected interactions with the protein export machinery (PTEX and exported protein-interacting complex) and a variety of proteins that either transit through the PV or reside on the parasite plasma membrane. Genetic knockdown or partial deletion of P113 did not significantly reduce parasite growth or protein export but did disrupt the morphology of the PVM, suggesting that P113 may play a role in maintaining normal PVM architecture.


Subject(s)
Malaria, Falciparum , Parasites , Animals , Erythrocytes/parasitology , Humans , Malaria, Falciparum/parasitology , Parasites/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Transport/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Vacuoles/metabolism
8.
Proc Natl Acad Sci U S A ; 119(13): e2115566119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35333655

ABSTRACT

SignificanceMitochondria are double-membraned eukaryotic organelles that house the proteins required for generation of ATP, the energy currency of cells. ATP generation within mitochondria is performed by five multisubunit complexes (complexes I to V), the assembly of which is an intricate process. Mutations in subunits of these complexes, or the suite of proteins that help them assemble, lead to a severe multisystem condition called mitochondrial disease. We show that SFXN4, a protein that causes mitochondrial disease when mutated, assists with the assembly of complex I. This finding explains why mutations in SFXN4 cause mitochondrial disease and is surprising because SFXN4 belongs to a family of amino acid transporter proteins, suggesting that it has undergone a dramatic shift in function through evolution.


Subject(s)
Electron Transport Complex I , Mitochondrial Diseases , Adenosine Triphosphate/metabolism , Electron Transport Complex I/metabolism , Humans , Membrane Proteins , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation
9.
J Cell Sci ; 134(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34313317

ABSTRACT

The mitochondrial inner membrane is a protein-rich environment containing large multimeric complexes, including complexes of the mitochondrial electron transport chain, mitochondrial translocases and quality control machineries. Although the inner membrane is highly proteinaceous, with 40-60% of all mitochondrial proteins localised to this compartment, little is known about the spatial distribution and organisation of complexes in this environment. We set out to survey the arrangement of inner membrane complexes using stochastic optical reconstruction microscopy (STORM). We reveal that subunits of the TIM23 complex, TIM23 and TIM44 (also known as TIMM23 and TIMM44, respectively), and the complex IV subunit COXIV, form organised clusters and show properties distinct from the outer membrane protein TOM20 (also known as TOMM20). Density based cluster analysis indicated a bimodal distribution of TIM44 that is distinct from TIM23, suggesting distinct TIM23 subcomplexes. COXIV is arranged in larger clusters that are disrupted upon disruption of complex IV assembly. Thus, STORM super-resolution microscopy is a powerful tool for examining the nanoscale distribution of mitochondrial inner membrane complexes, providing a 'visual' approach for obtaining pivotal information on how mitochondrial complexes exist in a cellular context.


Subject(s)
Mitochondria , Mitochondrial Membrane Transport Proteins , Animals , HEK293 Cells , HeLa Cells , Humans , Microscopy , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Protein Transport
10.
Mol Biol Cell ; 32(6): 475-491, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33476211

ABSTRACT

Acylglycerol kinase (AGK) is a mitochondrial lipid kinase that contributes to protein biogenesis as a subunit of the TIM22 complex at the inner mitochondrial membrane. Mutations in AGK cause Sengers syndrome, an autosomal recessive condition characterized by congenital cataracts, hypertrophic cardiomyopathy, skeletal myopathy, and lactic acidosis. We mapped the proteomic changes in Sengers patient fibroblasts and AGKKO cell lines to understand the effects of AGK dysfunction on mitochondria. This uncovered down-regulation of a number of proteins at the inner mitochondrial membrane, including many SLC25 carrier family proteins, which are predicted substrates of the complex. We also observed down-regulation of SFXN proteins, which contain five transmembrane domains, and show that they represent a novel class of TIM22 complex substrate. Perturbed biogenesis of SFXN proteins in cells lacking AGK reduces the proliferative capabilities of these cells in the absence of exogenous serine, suggesting that dysregulation of one-carbon metabolism is a molecular feature in the biology of Sengers syndrome.


Subject(s)
Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Carbon/metabolism , Carrier Proteins/metabolism , Cell Culture Techniques , Humans , MCF-7 Cells , Membrane Proteins/metabolism , Membrane Transport Proteins/physiology , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/physiology , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/physiology , Mutation , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Primary Cell Culture , Proteomics/methods
11.
FEBS Lett ; 595(8): 1107-1131, 2021 04.
Article in English | MEDLINE | ID: mdl-33314127

ABSTRACT

The majority of proteins localised to mitochondria are encoded by the nuclear genome, with approximately 1500 proteins imported into mammalian mitochondria. Dysfunction in this fundamental cellular process is linked to a variety of pathologies including neuropathies, cardiovascular disorders, myopathies, neurodegenerative diseases and cancer, demonstrating the importance of mitochondrial protein import machinery for cellular function. Correct import of proteins into mitochondria requires the co-ordinated activity of multimeric protein translocation and sorting machineries located in both the outer and inner mitochondrial membranes, directing the imported proteins to the destined mitochondrial compartment. This dynamic process maintains cellular homeostasis, and its dysregulation significantly affects cellular signalling pathways and metabolism. This review summarises current knowledge of the mammalian mitochondrial import machinery and the pathological consequences of mutation of its components. In addition, we will discuss the role of mitochondrial import in cancer, and our current understanding of the role of mitochondrial import in neurodegenerative diseases including Alzheimer's disease, Huntington's disease and Parkinson's disease.


Subject(s)
Mitochondria , Mitochondrial Diseases , Mitochondrial Proteins , Neoplasm Proteins , Neoplasms , Neurodegenerative Diseases , Animals , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Transport/genetics
12.
Mol Cell Proteomics ; 20: 100005, 2021.
Article in English | MEDLINE | ID: mdl-33177156

ABSTRACT

Modulation of the host cell is integral to the survival and replication of microbial pathogens. Several intracellular bacterial pathogens deliver bacterial proteins, termed "effector proteins" into the host cell during infection by sophisticated protein translocation systems, which manipulate cellular processes and functions. The functional contribution of individual effectors is poorly characterized, particularly in intracellular bacterial pathogens with large effector protein repertoires. Technical caveats have limited the capacity to study these proteins during a native infection, with many effector proteins having only been demonstrated to be translocated during over-expression of tagged versions. Here, we developed a novel strategy to examine effector proteins in the context of infection. We coupled a broad, unbiased proteomics-based screen with organelle purification to study the host-pathogen interactions occurring between the host cell mitochondrion and the Gram-negative, Q fever pathogen Coxiella burnetii. We identify four novel mitochondrially-targeted C. burnetii effector proteins, renamed Mitochondrial Coxiella effector protein (Mce) B to E. Examination of the subcellular localization of ectopically expressed proteins confirmed their mitochondrial localization, demonstrating the robustness of our approach. Subsequent biochemical analysis and affinity enrichment proteomics of one of these effector proteins, MceC, revealed the protein localizes to the inner membrane and can interact with components of the mitochondrial quality control machinery. Our study adapts high-sensitivity proteomics to study intracellular host-pathogen interactions, providing a robust strategy to examine the subcellular localization of effector proteins during native infection. This approach could be applied to a range of pathogens and host cell compartments to provide a rich map of effector dynamics throughout infection.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/physiology , Host-Pathogen Interactions , Mitochondria/metabolism , Mitochondria/microbiology , HEK293 Cells , HeLa Cells , Humans , Proteome , Proteomics , Q Fever , THP-1 Cells
14.
Elife ; 82019 11 04.
Article in English | MEDLINE | ID: mdl-31682224

ABSTRACT

Human Tim8a and Tim8b are members of an intermembrane space chaperone network, known as the small TIM family. Mutations in TIMM8A cause a neurodegenerative disease, Mohr-Tranebjærg syndrome (MTS), which is characterised by sensorineural hearing loss, dystonia and blindness. Nothing is known about the function of hTim8a in neuronal cells or how mutation of this protein leads to a neurodegenerative disease. We show that hTim8a is required for the assembly of Complex IV in neurons, which is mediated through a transient interaction with Complex IV assembly factors, in particular the copper chaperone COX17. Complex IV assembly defects resulting from loss of hTim8a leads to oxidative stress and changes to key apoptotic regulators, including cytochrome c, which primes cells for death. Alleviation of oxidative stress with Vitamin E treatment rescues cells from apoptotic vulnerability. We hypothesise that enhanced sensitivity of neuronal cells to apoptosis is the underlying mechanism of MTS.


Subject(s)
Deaf-Blind Disorders/physiopathology , Dystonia/physiopathology , Electron Transport Complex IV/metabolism , Intellectual Disability/physiopathology , Membrane Transport Proteins/metabolism , Neurons/metabolism , Optic Atrophy/physiopathology , Protein Multimerization , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Line , Copper Transport Proteins/metabolism , Humans , Membrane Transport Proteins/deficiency , Mitochondrial Precursor Protein Import Complex Proteins , Oxidative Stress , Protein Interaction Maps
15.
Front Immunol ; 9: 1809, 2018.
Article in English | MEDLINE | ID: mdl-30177930

ABSTRACT

FcγRIIa is an activating FcγR, unique to humans and non-human primates. It induces antibody-dependent proinflammatory responses and exists predominantly as FcγRIIa1. A unique splice variant, we designated FcγRIIa3, has been reported to be associated with anaphylactic reactions to intravenous immunoglobulins (IVIg) therapy. We aim to define the functional consequences of this FcγRIIa variant associated with adverse responses to IVIg therapy and evaluate the frequency of associated SNPs. FcγRIIa forms from macaque and human PBMCs were investigated for IgG-subclass specificity, biochemistry, membrane localization, and functional activity. Disease-associated SNPs were analyzed by sequencing genomic DNA from 224 individuals with immunodeficiency or autoimmune disease. FcγRIIa3 was identified in macaque and human PBMC. The FcγRIIa3 is distinguished from the canonical FcγRIIa1 by a unique 19-amino acid cytoplasmic insertion and these two FcγRIIa forms responded distinctly to antibody ligation. Whereas FcγRIIa1 was rapidly internalized, FcγRIIa3 was retained longer at the membrane, inducing greater calcium mobilization and cell degranulation. Four FCGR2A SNPs were identified including the previously reported intronic SNP associated with anaphylaxis, but in only 1 of 224 individuals. The unique cytoplasmic element of FcγRIIa3 delays internalization and is associated with enhanced cellular activation. The frequency of the immunodeficiency-associated SNP varies between disease populations but interestingly occurred at a lower frequency than previously reported. None-the-less enhanced FcγRIIa3 function may promote a proinflammatory environment and predispose to pathological inflammatory responses.


Subject(s)
Anaphylaxis/genetics , Anaphylaxis/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Anaphylaxis/diagnosis , Anaphylaxis/immunology , Animals , Biomarkers , Cell Degranulation/immunology , Disease Susceptibility , Fluorescent Antibody Technique , Gene Expression , Genetic Loci , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macaca , Mast Cells/immunology , Mast Cells/metabolism , Phenotype , Polymorphism, Single Nucleotide , Protein Binding , Protein Isoforms , Sequence Analysis, DNA
16.
Traffic ; 19(8): 605-623, 2018 08.
Article in English | MEDLINE | ID: mdl-29696751

ABSTRACT

Plasmodium falciparum, which causes malaria, extensively remodels its human host cells, particularly erythrocytes. Remodelling is essential for parasite survival by helping to avoid host immunity and assisting in the uptake of plasma nutrients to fuel rapid growth. Host cell renovation is carried out by hundreds of parasite effector proteins that are exported into the erythrocyte across an enveloping parasitophorous vacuole membrane (PVM). The Plasmodium translocon for exported (PTEX) proteins is thought to span the PVM and provide a channel that unfolds and extrudes proteins across the PVM into the erythrocyte. We show that exported reporter proteins containing mouse dihydrofolate reductase domains that inducibly resist unfolding become trapped at the parasite surface partly colocalizing with PTEX. When cargo is trapped, loop-like extensions appear at the PVM containing both trapped cargo and PTEX protein EXP2, but not additional components HSP101 and PTEX150. Following removal of the block-inducing compound, export of reporter proteins only partly recovers possibly because much of the trapped cargo is spatially segregated in the loop regions away from PTEX. This suggests that parasites have the means to isolate unfoldable cargo proteins from PTEX-containing export zones to avert disruption of protein export that would reduce parasite growth.


Subject(s)
Malaria, Falciparum/parasitology , Parasites/metabolism , Plasmodium falciparum/metabolism , Protein Transport/physiology , Protozoan Proteins/metabolism , Animals , Erythrocytes/parasitology , Humans , Malaria, Falciparum/blood , Mice , Vacuoles/parasitology
17.
J Cell Sci ; 129(11): 2170-81, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27076521

ABSTRACT

Cytosolic dynamin-related protein 1 (Drp1, also known as DNM1L) is required for both mitochondrial and peroxisomal fission. Drp1-dependent division of these organelles is facilitated by a number of adaptor proteins at mitochondrial and peroxisomal surfaces. To investigate the interplay of these adaptor proteins, we used gene-editing technology to create a suite of cell lines lacking the adaptors MiD49 (also known as MIEF2), MiD51 (also known as MIEF1), Mff and Fis1. Increased mitochondrial connectivity was observed following loss of individual adaptors, and this was further enhanced following the combined loss of MiD51 and Mff. Moreover, loss of adaptors also conferred increased resistance of cells to intrinsic apoptotic stimuli, with MiD49 and MiD51 showing the more prominent role. Using a proximity-based biotin labeling approach, we found close associations between MiD51, Mff and Drp1, but not Fis1. Furthermore, we found that MiD51 can suppress Mff-dependent enhancement of Drp1 GTPase activity. Our data indicates that Mff and MiD51 regulate Drp1 in specific ways to promote mitochondrial fission.


Subject(s)
Dynamins/metabolism , Membrane Proteins/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Death , Cell Line , Gene Editing , Mice, Inbred C57BL , Mice, Knockout , Peroxisomes/metabolism , Staining and Labeling
18.
J Cell Biol ; 204(4): 477-86, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24515348

ABSTRACT

Mitochondrial fission is important for organelle transport, inheritance, and turnover, and alterations in fission are seen in neurological disease. In mammals, mitochondrial fission is executed by dynamin-related protein 1 (Drp1), a cytosolic guanosine triphosphatase that polymerizes and constricts the organelle. Recruitment of Drp1 to mitochondria involves receptors including Mff, MiD49, and MiD51. MiD49/51 form foci at mitochondrial constriction sites and coassemble with Drp1 to drive fission. Here, we solved the crystal structure of the cytosolic domain of human MiD51, which adopts a nucleotidyltransferase fold. Although MiD51 lacks catalytic residues for transferase activity, it specifically binds guanosine diphosphate and adenosine diphosphate. MiD51 mutants unable to bind nucleotides were still able to recruit Drp1. Disruption of an additional region in MiD51 that is not part of the nucleotidyltransferase fold blocked Drp1 recruitment and assembly of MiD51 into foci. MiD51 foci are also dependent on the presence of Drp1, and after scission they are distributed to daughter organelles, supporting the involvement of MiD51 in the fission apparatus.


Subject(s)
Dynamins/physiology , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/metabolism , Adenosine Diphosphate/metabolism , Animals , Blotting, Western , Cells, Cultured , Crystallization , Crystallography, X-Ray , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Guanosine Diphosphate/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Peptide Elongation Factors/genetics
19.
Br J Pharmacol ; 171(8): 1870-89, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24117041

ABSTRACT

Progressive mitochondrial dysfunction is linked with the onset of many age-related pathologies and neurological disorders. Mitochondrial damage can come in many forms and be induced by a variety of cellular insults. To preserve organelle function during biogenesis or times of stress, multiple surveillance systems work to ensure the persistence of a functional mitochondrial network. This review provides an overview of these processes, which collectively contribute to the maintenance of a healthy mitochondrial population, which is critical for cell physiology and survival.


Subject(s)
Homeostasis , Mitochondrial Proteins/metabolism , Mitochondrial Turnover , Neurodegenerative Diseases/metabolism , Cell Death , Humans , Mitophagy , Models, Biological
20.
J Biol Chem ; 288(38): 27584-27593, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23921378

ABSTRACT

Drp1 (dynamin-related protein 1) is recruited to both mitochondrial and peroxisomal membranes to execute fission. Fis1 and Mff are Drp1 receptor/effector proteins of mitochondria and peroxisomes. Recently, MiD49 and MiD51 were also shown to recruit Drp1 to the mitochondrial surface; however, different reports have ascribed opposing roles in fission and fusion. Here, we show that MiD49 or MiD51 overexpression blocked fission by acting in a dominant-negative manner by sequestering Drp1 specifically at mitochondria, causing unopposed fusion events at mitochondria along with elongation of peroxisomes. Mitochondrial elongation caused by MiD49/51 overexpression required the action of fusion mediators mitofusins 1 and 2. Furthermore, at low level overexpression when MiD49 and MiD51 form discrete foci at mitochondria, mitochondrial fission events still occurred. Unlike Fis1 and Mff, MiD49 and MiD51 were not targeted to the peroxisomal surface, suggesting that they specifically act to facilitate Drp1-directed fission at mitochondria. Moreover, when MiD49 or MiD51 was targeted to the surface of peroxisomes or lysosomes, Drp1 was specifically recruited to these organelles. Moreover, the Drp1 recruitment activity of MiD49/51 appeared stronger than that of Mff or Fis1. We conclude that MiD49 and MiD51 can act independently of Mff and Fis1 in Drp1 recruitment and suggest that they provide specificity to the division of mitochondria.


Subject(s)
Dynamins/metabolism , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Peptide Elongation Factors/metabolism , Animals , Dynamins/genetics , GTP Phosphohydrolases/genetics , HeLa Cells , Humans , Lysosomes/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Peptide Elongation Factors/genetics , Peroxisomes/genetics , Peroxisomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...