Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Methods Mol Biol ; 2615: 173-188, 2023.
Article in English | MEDLINE | ID: mdl-36807792

ABSTRACT

Reminiscent of their evolutionary origin, mitochondria contain their own genome (mtDNA) compacted into the mitochondrial chromosome or nucleoid (mt-nucleoid). Many mitochondrial disorders are characterized by disruption of mt-nucleoids, either by direct mutation of genes involved in mtDNA organization or by interfering with other vital proteins for mitochondrial function. Thus, changes in mt-nucleoid morphology, distribution, and structure are a common feature in many human diseases and can be exploited as an indicator of cellular fitness. Electron microscopy provides the highest possible resolution that can be achieved, delivering spatial and structural information about all cellular structures. Recently, the ascorbate peroxidase APEX2 has been used to increase transmission electron microscopy (TEM) contrast by inducing diaminobenzidine (DAB) precipitation. DAB has the ability to accumulate osmium during classical EM sample preparation and, due to its high electron density, provides strong contrast for TEM. Among the nucleoid proteins, the mitochondrial helicase Twinkle fused with APEX2 has been successfully used to target mt-nucleoids, providing a tool to visualize these subcellular structures with high contrast and with the resolution of an electron microscope. In the presence of H2O2, APEX2 catalyzes the polymerization of DAB, generating a brown precipitate that can be visualized in specific regions of the mitochondrial matrix. Here, we provide a detailed protocol to generate murine cell lines expressing a transgenic variant of Twinkle, suitable to target and visualize mt-nucleoids. We also describe all the necessary steps to validate the cell lines prior to electron microscopy imaging and offer examples of anticipated results.


Subject(s)
Hydrogen Peroxide , Mitochondria , Animals , Mice , Humans , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , DNA, Mitochondrial/genetics , Animals, Genetically Modified , DNA Helicases/metabolism , Microscopy, Electron, Transmission , Mitochondrial Proteins/metabolism , Endonucleases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Multifunctional Enzymes
2.
Nat Commun ; 13(1): 6704, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36344526

ABSTRACT

Understanding the mechanisms governing selective turnover of mutation-bearing mtDNA is fundamental to design therapeutic strategies against mtDNA diseases. Here, we show that specific mtDNA damage leads to an exacerbated mtDNA turnover, independent of canonical macroautophagy, but relying on lysosomal function and ATG5. Using proximity labeling and Twinkle as a nucleoid marker, we demonstrate that mtDNA damage induces membrane remodeling and endosomal recruitment in close proximity to mitochondrial nucleoid sub-compartments. Targeting of mitochondrial nucleoids is controlled by the ATAD3-SAMM50 axis, which is disrupted upon mtDNA damage. SAMM50 acts as a gatekeeper, influencing BAK clustering, controlling nucleoid release and facilitating transfer to endosomes. Here, VPS35 mediates maturation of early endosomes to late autophagy vesicles where degradation occurs. In addition, using a mouse model where mtDNA alterations cause impairment of muscle regeneration, we show that stimulation of lysosomal activity by rapamycin, selectively removes mtDNA deletions without affecting mtDNA copy number, ameliorating mitochondrial dysfunction. Taken together, our data demonstrates that upon mtDNA damage, mitochondrial nucleoids are eliminated outside the mitochondrial network through an endosomal-mitophagy pathway. With these results, we unveil the molecular players of a complex mechanism with multiple potential benefits to understand mtDNA related diseases, inherited, acquired or due to normal ageing.


Subject(s)
DNA, Mitochondrial , Mitochondrial Membranes , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitophagy
3.
Front Cell Dev Biol ; 9: 698503, 2021.
Article in English | MEDLINE | ID: mdl-34395429

ABSTRACT

CD30, a member of the TNF receptor superfamily, is selectively expressed on a subset of activated lymphocytes and on malignant cells of certain lymphomas, such as classical Hodgkin Lymphoma (cHL), where it activates critical bystander cells in the tumor microenvironment. Therefore, it is not surprising that the CD30 antibody-drug conjugate Brentuximab Vedotin (BV) represents a powerful, FDA-approved treatment option for CD30+ hematological malignancies. However, BV also exerts a strong anti-cancer efficacy in many cases of diffuse large B cell lymphoma (DLBCL) with poor CD30 expression, even when lacking detectable CD30+ tumor cells. The mechanism remains enigmatic. Because CD30 is released on extracellular vesicles (EVs) from both, malignant and activated lymphocytes, we studied whether EV-associated CD30 might end up in CD30- tumor cells to provide binding sites for BV. Notably, CD30+ EVs bind to various DLBCL cell lines as well as to the FITC-labeled variant of the antibody-drug conjugate BV, thus potentially conferring the BV binding also to CD30- cells. Confocal microscopy and imaging cytometry studies revealed that BV binding and uptake depend on CD30+ EVs. Since BV is only toxic toward CD30- DLBCL cells when CD30+ EVs support its uptake, we conclude that EVs not only communicate within the tumor microenvironment but also influence cancer treatment. Ultimately, the CD30-based BV not only targets CD30+ tumor cell but also CD30- DLBCL cells in the presence of CD30+ EVs. Our study thus provides a feasible explanation for the clinical impact of BV in CD30- DLBCL and warrants confirming studies in animal models.

4.
J Extracell Vesicles ; 8(1): 1596016, 2019.
Article in English | MEDLINE | ID: mdl-30988894

ABSTRACT

The expanding field of extracellular vesicle (EV) research needs reproducible and accurate methods to characterize single EVs. Nanoparticle Tracking Analysis (NTA) is commonly used to determine EV concentration and diameter. As the EV field is lacking methods to easily confirm and validate NTA data, questioning the reliability of measurements remains highly important. In this regard, a comparison addressing measurement quality between different NTA devices such as Malvern's NanoSight NS300 or Particle Metrix' ZetaView has not yet been conducted. To evaluate the accuracy and repeatability of size and concentration determinations of both devices, we employed comparative methods including transmission electron microscopy (TEM) and single particle interferometric reflectance imaging sensing (SP-IRIS) by ExoView. Multiple test measurements with nanospheres, liposomes and ultracentrifuged EVs from human serum and cell culture supernatant were performed. Additionally, serial dilutions and freeze-thaw cycle-dependent EV decrease were measured to determine the robustness of each system. Strikingly, NanoSight NS300 exhibited a 2.0-2.1-fold overestimation of polystyrene and silica nanosphere concentration. By measuring serial dilutions of EV samples, we demonstrated higher accuracy in concentration determination by ZetaView (% BIAS range: 2.7-8.5) in comparison with NanoSight NS300 (% BIAS range: 32.9-36.8). The concentration measurements by ZetaView were also more precise (% CV range: 0.0-4.7) than measurements by NanoSight NS300 (% CV range: 5.4-10.7). On the contrary, quantitative TEM imaging indicated more accurate EV sizing by NanoSight NS300 (% DTEM range: 79.5-134.3) compared to ZetaView (% DTEM range: 111.8-205.7), while being equally repeatable (NanoSight NS300% CV range: 0.8-6.7; ZetaView: 1.4-7.8). However, both devices failed to report a peak EV diameter below 60 nm compared to TEM and SP-IRIS. Taken together, NTA devices differ strongly in their hardware and software affecting measuring results. ZetaView provided a more accurate and repeatable depiction of EV concentration, whereas NanoSight NS300 supplied size measurements of higher resolution.

5.
Mycoses ; 62(7): 562-569, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31034691

ABSTRACT

Invasive mould infections (IMI) in immunocompromised patients are difficult to diagnose. Early and targeted treatment is paramount, but minimally invasive tests reliably identifying pathogens are lacking. We previously showed that monitoring pathogen-specific CD4+T cells in peripheral blood using upregulation of induced CD154 positive lymphocytes can be used to diagnose acute IMI. Here, we validate our findings in an independent patient cohort. We stimulated peripheral blood cells from at-risk patients with Aspergillus spp. and Mucorales lysates and quantitated mould-reactive CD4/CD69/CD154 positive lymphocytes via flow cytometry. Mould-reactive lymphocytes were quantitated in 115 at-risk patients. In 38 (33%) patients, the test was not evaluable, mainly due to low T cell counts or non-reactive positive control. Test results were evaluable in 77 (67%) patients. Of these, four patients (5%) had proven IMI and elevated mould-reactive T cell signals. Of 73 (95%) patients without proven IMI, 59 (81%) had mould-reactive T cell signals within normal range. Fourteen (19%) patients without confirmed IMI showed elevated T cell signals and 11 of those received antifungal treatment. The mould-reactive lymphocyte assay identified presence of IMI with a sensitivity of 100% and specificity of 81%. The mould-reactive lymphocyte assay correctly identified all patients with proven IMI. Assay applicability is limited by low T cell counts during bone marrow suppression. The assay has the potential to support diagnosis of invasive mould infection to facilitate tailored treatment even when biopsies are contraindicated or cultures remain negative.


Subject(s)
Aspergillus/immunology , CD4-Positive T-Lymphocytes/immunology , Invasive Fungal Infections/diagnosis , Mucorales/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Aged , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD4-Positive T-Lymphocytes/chemistry , CD40 Ligand/analysis , Female , Flow Cytometry , Humans , Immunocompromised Host , Lectins, C-Type/analysis , Male , Middle Aged , Prospective Studies , Sensitivity and Specificity , T-Lymphocyte Subsets/chemistry , Young Adult
6.
Sci Rep ; 9(1): 2069, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765836

ABSTRACT

MicroRNAs (miRNAs) are small non-coding nucleotides playing a crucial role in posttranscriptional expression and regulation of target genes in nearly all kinds of cells. In this study, we demonstrate a reliable and efficient capture and purification of miRNAs and intracellular proteins using magnetic nanoparticles functionalized with antisense oligonucleotides. For this purpose, a tumor suppressor miRNA (miR-198), deregulated in several human cancer types, was chosen as the model oligonucleotide. Magnetite nanoparticles carrying the complementary sequence of miR-198 (miR-198 antisense) on their surface were delivered into cells and subsequently used for the extracellular transport of miRNA and proteins. The successful capture of miR-198 was demonstrated by isolating RNA from magnetic nanoparticles followed by real-time PCR quantification. Our experimental data showed that antisense-coated particles captured 5-fold higher amounts of miR-198 when compared to the control nanoparticles. Moreover, several proteins that could play a significant role in miR-198 biogenesis were found attached to miR-198 conjugated nanoparticles and analyzed by mass spectrometry. Our findings demonstrate that a purpose-driven vectorization of magnetic nanobeads with target-specific recognition ligands is highly efficient in selectively transporting miRNA and disease-relevant proteins out of cells and could become a reliable and useful tool for future diagnostic, therapeutic and analytical applications.


Subject(s)
MicroRNAs/genetics , Oligonucleotides, Antisense/genetics , Proteins/genetics , Cell Line , Humans , Magnetic Phenomena
7.
Front Microbiol ; 9: 1381, 2018.
Article in English | MEDLINE | ID: mdl-29988394

ABSTRACT

Background: Blood or tissue culture or histology prove invasive Candida infection, but long time to result, limited feasibility and sensitivity call for new approaches. In this pilot project, we describe the diagnostic potential of quantitating Candida-reactive, CD4/CD69/CD154 positive lymphocytes in blood of patients with invasive Candida infection. Methods: We used flow cytometry quantitating Candida-reactive, CD4/CD69/CD154 positive lymphocytes from peripheral blood of patients with invasive Candida infection, from patients at risk and healthy volunteers as controls. Results: Elevated levels of Candida-reactive lymphocytes were measured in 13 patients with proven invasive Candida infection and in one patient with probable hepatosplenic candidiasis. Results of three candidemia patients were uninterpretable due to autofluorescence of samples. Twelve of 13 patients had Candida identified to species level by conventional methods, and T cell reactivity correctly identified Candida species in 10 of 12 patients. Nine hematological high-risk patients and 14 healthy donors had no elevated Candida-reactive T cell counts. Conclusions: This Candida-reactive lymphocyte assay correctly identified the majority of patients with invasive Candida infection and the respective species. Our assay has the potential to support diagnosis of invasive Candida infection to species level and to facilitate tailored treatment even when biopsies are contraindicated or cultures remain negative.

8.
Nat Commun ; 8(1): 1250, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29093447

ABSTRACT

Generation of a barrier in multi-layered epithelia like the epidermis requires restricted positioning of functional tight junctions (TJ) to the most suprabasal viable layer. This positioning necessitates tissue-level polarization of junctions and the cytoskeleton through unknown mechanisms. Using quantitative whole-mount imaging, genetic ablation, and traction force microscopy and atomic force microscopy, we find that ubiquitously localized E-cadherin coordinates tissue polarization of tension-bearing adherens junction (AJ) and F-actin organization to allow formation of an apical TJ network only in the uppermost viable layer. Molecularly, E-cadherin localizes and tunes EGFR activity and junctional tension to inhibit premature TJ complex formation in lower layers while promoting increased tension and TJ stability in the granular layer 2. In conclusion, our data identify an E-cadherin-dependent mechanical circuit that integrates adhesion, contractile forces and biochemical signaling to drive the polarized organization of junctional tension necessary to build an in vivo epithelial barrier.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Epidermis/metabolism , ErbB Receptors/metabolism , Mechanotransduction, Cellular , Tight Junctions/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Adherens Junctions/ultrastructure , Animals , Cell Differentiation , Cell Proliferation , Epidermis/ultrastructure , Mice , Mice, Knockout , Microscopy, Atomic Force , Signal Transduction , Tight Junctions/ultrastructure
9.
EMBO Rep ; 18(9): 1521-1535, 2017 09.
Article in English | MEDLINE | ID: mdl-28710093

ABSTRACT

Primary cilia are sensory, antennae-like organelles present on the surface of many cell types. They have been involved in a variety of diseases collectively termed ciliopathies. As cilia are essential regulators of cell signaling, the composition of the ciliary membrane needs to be strictly regulated. To understand regulatory processes at the ciliary membrane, we report the targeting of a genetically engineered enzyme specifically to the ciliary membrane to allow biotinylation and identification of the membrane-associated proteome. Bioinformatic analysis of the comprehensive dataset reveals high-stoichiometric presence of actin-binding proteins inside the cilium. Immunofluorescence stainings and complementary interaction proteomic analyses confirm these findings. Depolymerization of branched F-actin causes further enrichment of the actin-binding and actin-related proteins in cilia, including Myosin 5a (Myo5a). Interestingly, Myo5a knockout decreases ciliation while enhanced levels of Myo5a are observed in cilia upon induction of ciliary disassembly. In summary, we present a novel approach to investigate dynamics of the ciliary membrane proteome in mammalian cells and identify actin-binding proteins as mechanosensitive components of cilia that might have important functions in cilia membrane dynamics.


Subject(s)
Actins/metabolism , Cilia/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Proteome/metabolism , Actins/chemistry , Animals , Computational Biology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Membranes/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Myosins/deficiency , Myosins/genetics , Myosins/metabolism , Proteomics , Signal Transduction
10.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L687-L698, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28684544

ABSTRACT

Deficiency of the extracellular matrix protein latent transforming growth factor-ß (TGF-ß)-binding protein-4 (LTBP4) results in lack of intact elastic fibers, which leads to disturbed pulmonary development and lack of normal alveolarization in humans and mice. Formation of alveoli and alveolar septation in pulmonary development requires the concerted interaction of extracellular matrix proteins, growth factors such as TGF-ß, fibroblasts, and myofibroblasts to promote elastogenesis as well as vascular formation in the alveolar septae. To investigate the role of LTBP4 in this context, lungs of LTBP4-deficient (Ltbp4-/-) mice were analyzed in close detail. We elucidate the role of LTBP4 in pulmonary alveolarization and show that three different, interacting mechanisms might contribute to alveolar septation defects in Ltbp4-/- lungs: 1) absence of an intact elastic fiber network, 2) reduced angiogenesis, and 3) upregulation of TGF-ß activity resulting in profibrotic processes in the lung.


Subject(s)
Elastic Tissue/pathology , Fibroblasts/pathology , Fibrosis/pathology , Latent TGF-beta Binding Proteins/physiology , Lung/pathology , Neovascularization, Pathologic/pathology , Pulmonary Alveoli/pathology , Animals , Cells, Cultured , Elastic Tissue/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Lung/blood supply , Lung/metabolism , Male , Mice , Mice, Knockout , Neovascularization, Pathologic/metabolism , Organogenesis/physiology , Pulmonary Alveoli/metabolism , Transforming Growth Factor beta/metabolism
11.
Proc Natl Acad Sci U S A ; 114(28): E5559-E5568, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28645896

ABSTRACT

Dynamin-like proteins (DLPs) mediate various membrane fusion and fission processes within the cell, which often require the polymerization of DLPs. An IFN-inducible family of DLPs, the guanylate-binding proteins (GBPs), is involved in antimicrobial and antiviral responses within the cell. Human guanylate-binding protein 1 (hGBP1), the founding member of GBPs, is also engaged in the regulation of cell adhesion and migration. Here, we show how the GTPase cycle of farnesylated hGBP1 (hGBP1F) regulates its self-assembly and membrane interaction. Using vesicles of various sizes as a lipid bilayer model, we show GTP-dependent membrane binding of hGBP1F In addition, we demonstrate nucleotide-dependent tethering ability of hGBP1F Furthermore, we report nucleotide-dependent polymerization of hGBP1F, which competes with membrane binding of the protein. Our results show that hGBP1F acts as a nucleotide-controlled molecular switch by modulating the accessibility of its farnesyl moiety, which does not require any supportive proteins.


Subject(s)
GTP-Binding Proteins/metabolism , Guanosine Triphosphate/chemistry , Polymers/chemistry , Binding Sites , Catalysis , Cell Membrane/metabolism , GTP Phosphohydrolases/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HeLa Cells , Humans , Hydrolysis , Immunity, Innate , Liposomes/chemistry , Microscopy, Electron , Polymerization , Prenylation , Protein Binding
12.
J Cell Biol ; 216(3): 675-693, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28188211

ABSTRACT

Mitochondria are essential organelles that host crucial metabolic pathways and produce adenosine triphosphate. The mitochondrial proteome is heterogeneous among tissues and can dynamically change in response to different metabolic conditions. Although the transcriptional programs that govern mitochondrial biogenesis and respiratory function are well known, posttranscriptional regulatory mechanisms remain unclear. In this study, we show that the cytosolic RNA-binding protein clustered mitochondria homologue (CLUH) regulates the expression of a mitochondrial protein network supporting key metabolic programs required under nutrient deprivation. CLUH exerts its function by controlling the stability and translation of target messenger RNAs. In the absence of Cluh, mitochondria are severely depleted of crucial enzymes involved in catabolic energy-converting pathways. CLUH preserves oxidative mitochondrial function and glucose homeostasis, thus preventing death at the fetal-neonatal transition. In the adult liver, CLUH ensures maximal respiration capacity and the metabolic response to starvation. Our results shed new light on the posttranscriptional mechanisms controlling the expression of mitochondrial proteins and suggest novel strategies to tailor mitochondrial function to physiological and pathological conditions.


Subject(s)
Mitochondria/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cytosol/metabolism , Cytosol/physiology , Energy Metabolism/physiology , Gene Expression Regulation/physiology , Homeostasis/physiology , Metabolism/physiology , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , RNA Interference/physiology
13.
EMBO Rep ; 16(6): 719-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25825408

ABSTRACT

In this study, we show for the first time that the therapeutic antagonization of inhibitor of apoptosis proteins (IAPs) inhibits B16 melanoma growth by disrupting tumor vasculature. Specifically, the treatment of mice bearing B16 melanoma with an IAP antagonist compound A (Comp A) inhibits tumor growth not by inducing direct cytotoxicity against B16 cells but rather by a hitherto unrecognized antiangiogenic activity against tumor vessels. Our detailed analysis showed that Comp A treatment induces NF-κB activity in B16 tumor cells and facilitates the production of TNF. In the presence of Comp A, endothelial cells (ECs) become highly susceptible to TNF and undergo apoptotic cell death. Accordingly, the antiangiogenic and growth-attenuating effects of Comp A treatment were completely abolished in TNF-R knockout mice. This novel targeting approach could be of clinical value in controlling pathological neoangiogenesis under inflammatory condition while sparing blood vessels under normal condition.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/pathology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Animals , Apoptosis/drug effects , Inflammation/physiopathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Neovascularization, Pathologic , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
EMBO J ; 33(9): 1011-26, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24681487

ABSTRACT

The m-AAA protease subunit AFG3L2 is involved in degradation and processing of substrates in the inner mitochondrial membrane. Mutations in AFG3L2 are associated with spinocerebellar ataxia SCA28 in humans and impair axonal development and neuronal survival in mice. The loss of AFG3L2 causes fragmentation of the mitochondrial network. However, the pathogenic mechanism of neurodegeneration in the absence of AFG3L2 is still unclear. Here, we show that depletion of AFG3L2 leads to a specific defect of anterograde transport of mitochondria in murine cortical neurons. We observe similar transport deficiencies upon loss of AFG3L2 in OMA1-deficient neurons, indicating that they are not caused by OMA1-mediated degradation of the dynamin-like GTPase OPA1 and inhibition of mitochondrial fusion. Treatment of neurons with antioxidants, such as N-acetylcysteine or vitamin E, or decreasing tau levels in axons restored mitochondrial transport in AFG3L2-depleted neurons. Consistently, tau hyperphosphorylation and activation of ERK kinases are detected in mouse neurons postnatally deleted for Afg3l2. We propose that reactive oxygen species signaling leads to cytoskeletal modifications that impair mitochondrial transport in neurons lacking AFG3L2.


Subject(s)
ATP-Dependent Proteases/genetics , Mitochondria/metabolism , tau Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Acetylcysteine/pharmacology , Animals , Biological Transport/drug effects , Biological Transport/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryo, Mammalian , MAP Kinase Signaling System/genetics , Metalloproteases/genetics , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphorylation/genetics , Reactive Oxygen Species/pharmacology
15.
J Cell Biol ; 204(6): 919-29, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24616225

ABSTRACT

Mitochondrial fusion and structure depend on the dynamin-like GTPase OPA1, whose activity is regulated by proteolytic processing. Constitutive OPA1 cleavage by YME1L and OMA1 at two distinct sites leads to the accumulation of both long and short forms of OPA1 and maintains mitochondrial fusion. Stress-induced OPA1 processing by OMA1 converts OPA1 completely into short isoforms, inhibits fusion, and triggers mitochondrial fragmentation. Here, we have analyzed the function of different OPA1 forms in cells lacking YME1L, OMA1, or both. Unexpectedly, deletion of Oma1 restored mitochondrial tubulation, cristae morphogenesis, and apoptotic resistance in cells lacking YME1L. Long OPA1 forms were sufficient to mediate mitochondrial fusion in these cells. Expression of short OPA1 forms promoted mitochondrial fragmentation, which indicates that they are associated with fission. Consistently, GTPase-inactive, short OPA1 forms partially colocalize with ER-mitochondria contact sites and the mitochondrial fission machinery. Thus, OPA1 processing is dispensable for fusion but coordinates the dynamic behavior of mitochondria and is crucial for mitochondrial integrity and quality control.


Subject(s)
GTP Phosphohydrolases/metabolism , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Animals , Apoptosis , HEK293 Cells , Humans , Metalloendopeptidases/genetics , Metalloproteases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Size , Organelle Shape , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport
16.
Plant Physiol ; 165(1): 186-95, 2014 May.
Article in English | MEDLINE | ID: mdl-24676857

ABSTRACT

In Arabidopsis (Arabidopsis thaliana), root hairs are formed in cell files over the cleft of underlying cortex cells. This pattern is established by a well-known gene regulatory network of transcription factors. In this study, we show that WRKY75 suppresses root hair development in nonroot hair files and that it represses the expression of TRIPTYCHON and CAPRICE. The WRKY75 protein binds to the CAPRICE promoter in a yeast one-hybrid assay. Binding to the promoter fragment requires an intact WRKY protein-binding motif, the W box. A comparison of the spatial expression of WRKY75 and the localization of the WRKY75 protein revealed that WRKY75 is expressed in the pericycle and vascular tissue and that the WRKY75 RNA or protein moves into the epidermis.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Body Patterning/genetics , Gene Expression Regulation, Plant , Genes, Plant , Plant Roots/growth & development , Plant Roots/genetics , Transcription Factors/metabolism , Arabidopsis/growth & development , Base Sequence , Glucuronidase/metabolism , Molecular Sequence Data , Mutation/genetics , Phenotype , Plant Epidermis/cytology , Plant Epidermis/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , RNA Transport , RNA, Plant/metabolism , Transcription, Genetic , Two-Hybrid System Techniques
17.
Mol Cell Biol ; 31(16): 3241-51, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21690291

ABSTRACT

Neph proteins are evolutionarily conserved membrane proteins of the immunoglobulin superfamily that control the formation of specific intercellular contacts. Cell recognition through these proteins is essential in diverse cellular contexts such as patterning of the compound eye in Drosophila melanogaster, neuronal connectivity in Caenorhabditis elegans, and the formation of the kidney filtration barrier in mammals. Here we identify the PDZ and BAR domain protein PICK1 (protein interacting with C-kinase 1) as a Neph-interacting protein. Binding required dimerization of PICK1, was dependent on PDZ domain protein interactions, and mediated stabilization of Neph1 at the plasma membrane. Moreover, protein kinase C (PKCα) activity facilitated the interaction through releasing Neph proteins from their binding to the multidomain scaffolding protein zonula occludens 1 (ZO-1), another PDZ domain protein. In Drosophila, the Neph homologue Roughest is essential for sorting of interommatidial precursor cells and patterning of the compound eye. RNA interference-mediated knockdown of PICK1 in the Drosophila eye imaginal disc caused a Roughest destabilization at the plasma membrane and a phenotype that resembled rst mutation. These data indicate that Neph proteins and PICK1 synergistically regulate cell recognition and contact formation.


Subject(s)
Carrier Proteins/physiology , Cell Communication , Membrane Proteins/metabolism , Morphogenesis , Nuclear Proteins/physiology , Animals , Drosophila , Drosophila melanogaster , Eye/cytology , Humans , PDZ Domains , Protein Binding , Protein Kinase C , Protein Multimerization , Protein Stability , Protein Structure, Tertiary
18.
BMC Biol ; 8: 100, 2010 Jul 26.
Article in English | MEDLINE | ID: mdl-20659315

ABSTRACT

BACKGROUND: Mitochondria are highly dynamic organelles whose morphology and position within the cell is tightly coupled to metabolic function. There is a limited list of essential proteins that regulate mitochondrial morphology and the mechanisms that govern mitochondrial dynamics are poorly understood. However, recent evidence indicates that the core machinery that governs mitochondrial dynamics is linked within complex intracellular signalling cascades, including apoptotic pathways, cell cycle transitions and nuclear factor kappa B activation. Given the emerging importance of mitochondrial plasticity in cell signalling pathways and metabolism, it is essential that we develop tools to quantitatively analyse the processes of fission and fusion. In terms of mitochondrial fusion, the field currently relies upon on semi-quantitative assays which, even under optimal conditions, are labour-intensive, low-throughput and require complex imaging techniques. RESULTS: In order to overcome these technical limitations, we have developed a new, highly quantitative cell-free assay for mitochondrial fusion in mammalian cells. This assay system has allowed us to establish the energetic requirements for mitochondrial fusion. In addition, our data reveal a dependence on active protein phosphorylation for mitochondrial fusion, confirming emerging evidence that mitochondrial fusion is tightly integrated within the global cellular response to signaling events. Indeed, we have shown that cytosol derived from cells stimulated with different triggers either enhance or inhibit the cell-free fusion reaction. CONCLUSIONS: The adaptation of this system to high-throughput analysis will provide an unprecedented opportunity to identify and characterize novel regulatory factors. In addition, it provides a framework for a detailed mechanistic analysis of the process of mitochondrial fusion and the various axis of regulation that impinge upon this process in a wide range of cellular conditions.See Commentary: http://www.biomedcentral.com/1741-7007/8/99.


Subject(s)
Biochemistry/methods , Membrane Fusion , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , HeLa Cells , Humans , Mitochondria/ultrastructure , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/metabolism , Nucleotides/metabolism
19.
Proc Natl Acad Sci U S A ; 107(13): 6028-33, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231445

ABSTRACT

c-Jun N-terminal kinase (JNK) 1-dependent signaling plays a crucial role in the development of obesity-associated insulin resistance. Here we demonstrate that JNK activation not only occurs in peripheral tissues, but also in the hypothalamus and pituitary of obese mice. To resolve the importance of JNK1 signaling in the hypothalamic/pituitary circuitry, we have generated mice with a conditional inactivation of JNK1 in nestin-expressing cells (JNK1(DeltaNES) mice). JNK1(DeltaNES) mice exhibit improved insulin sensitivity both in the CNS and in peripheral tissues, improved glucose metabolism, as well as protection from hepatic steatosis and adipose tissue dysfunction upon high-fat feeding. Moreover, JNK1(DeltaNES) mice also show reduced somatic growth in the presence of reduced circulating growth hormone (GH) and insulin-like growth factor 1 (IGF1) concentrations, as well as increased thyroid axis activity. Collectively, these experiments reveal an unexpected, critical role for hypothalamic/pituitary JNK1 signaling in the coordination of metabolic/endocrine homeostasis.


Subject(s)
Glucose/metabolism , Hypothalamus/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Pituitary Gland/metabolism , Adiposity/physiology , Animals , Body Weight/physiology , Dietary Fats/administration & dosage , Growth Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Intermediate Filament Proteins/metabolism , Mice , Mice, Obese , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Nerve Tissue Proteins/metabolism , Nestin , Signal Transduction , Thyroid Gland/metabolism
20.
Curr Biol ; 18(2): 102-8, 2008 Jan 22.
Article in English | MEDLINE | ID: mdl-18207745

ABSTRACT

Mitochondria and peroxisomes share a number of common biochemical processes, including the beta oxidation of fatty acids and the scavenging of peroxides. Here, we identify a new outer-membrane mitochondria-anchored protein ligase (MAPL) containing a really interesting new gene (RING)-finger domain. Overexpression of MAPL leads to mitochondrial fragmentation, indicating a regulatory function controlling mitochondrial morphology. In addition, confocal- and electron-microscopy studies of MAPL-YFP led to the observation that MAPL is also incorporated within unique, DRP1-independent, 70-100 nm diameter mitochondria-derived vesicles (MDVs). Importantly, vesicles containing MAPL exclude another outer-membrane marker, TOM20, and vesicles containing TOM20 exclude MAPL, indicating that MDVs selectively incorporate their cargo. We further demonstrate that MAPL-containing vesicles fuse with a subset of peroxisomes, marking the first evidence for a direct relationship between these two functionally related organelles. In contrast, a distinct vesicle population labeled with TOM20 does not fuse with peroxisomes, indicating that the incorporation of specific cargo is a primary determinant of MDV fate. These data are the first to identify MAPL, describe and characterize MDVs, and define a new intracellular transport route between mitochondria and peroxisomes.


Subject(s)
Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Peroxisomes/metabolism , Transcription Factors/metabolism , Transport Vesicles/metabolism , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Mitochondria/metabolism , RING Finger Domains/physiology , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...