38 results on '"Boris Reljic"'
Search Results
2. Applying Sodium Carbonate Extraction Mass Spectrometry to Investigate Defects in the Mitochondrial Respiratory Chain
- Author
-
David R. L. Robinson, Daniella H. Hock, Linden Muellner-Wong, Roopasingam Kugapreethan, Boris Reljic, Elliot E. Surgenor, Carlos H. M. Rodrigues, Nikeisha J. Caruana, and David A. Stroud
- Subjects
mitochondria ,proteomic analyses ,membrane protein ,OXPHOS (oxidative phosphorylation) ,respiratory chain assembly ,carbonate extraction ,Biology (General) ,QH301-705.5 - Abstract
Mitochondria are complex organelles containing 13 proteins encoded by mitochondrial DNA and over 1,000 proteins encoded on nuclear DNA. Many mitochondrial proteins are associated with the inner or outer mitochondrial membranes, either peripherally or as integral membrane proteins, while others reside in either of the two soluble mitochondrial compartments, the mitochondrial matrix and the intermembrane space. The biogenesis of the five complexes of the oxidative phosphorylation system are exemplars of this complexity. These large multi-subunit complexes are comprised of more than 80 proteins with both membrane integral and peripheral associations and require soluble, membrane integral and peripherally associated assembly factor proteins for their biogenesis. Mutations causing human mitochondrial disease can lead to defective complex assembly due to the loss or altered function of the affected protein and subsequent destabilization of its interactors. Here we couple sodium carbonate extraction with quantitative mass spectrometry (SCE-MS) to track changes in the membrane association of the mitochondrial proteome across multiple human knockout cell lines. In addition to identifying the membrane association status of over 840 human mitochondrial proteins, we show how SCE-MS can be used to understand the impacts of defective complex assembly on protein solubility, giving insights into how specific subunits and sub-complexes become destabilized.
- Published
- 2022
- Full Text
- View/download PDF
3. Distinct initiating events underpin the immune and metabolic heterogeneity of KRAS-mutant lung adenocarcinoma
- Author
-
Sarah A. Best, Sheryl Ding, Ariena Kersbergen, Xueyi Dong, Ji-Ying Song, Yi Xie, Boris Reljic, Kaiming Li, James E. Vince, Vivek Rathi, Gavin M. Wright, Matthew E. Ritchie, and Kate D. Sutherland
- Subjects
Science - Abstract
Lung adenocarcinomas frequently harbour KRAS mutations, of which a subset are characterized by co-mutation of KEAP1. Here the authors show, in mice, that Kras G12D mutant tumours are metabolically distinct, with a bronchiolar cell-of-origin.
- Published
- 2019
- Full Text
- View/download PDF
4. Metabolic remodeling of dystrophic skeletal muscle reveals biological roles for dystrophin and utrophin in adaptation and plasticity
- Author
-
Justin P. Hardee, Karen J.B. Martins, Paula M. Miotto, James G. Ryall, Stefan M. Gehrig, Boris Reljic, Timur Naim, Jin D. Chung, Jen Trieu, Kristy Swiderski, Ashleigh M. Philp, Andrew Philp, Matthew J. Watt, David A. Stroud, Rene Koopman, Gregory R. Steinberg, and Gordon S. Lynch
- Subjects
Muscular dystrophy ,Muscle adaptation ,Dystrophin ,Utrophin ,Oxidative metabolism ,Internal medicine ,RC31-1245 - Abstract
Objectives: Preferential damage to fast, glycolytic myofibers is common in many muscle-wasting diseases, including Duchenne muscular dystrophy (DMD). Promoting an oxidative phenotype could protect muscles from damage and ameliorate the dystrophic pathology with therapeutic relevance, but developing efficacious strategies requires understanding currently unknown biological roles for dystrophin and utrophin in dystrophic muscle adaptation and plasticity. Methods: Combining whole transcriptome RNA sequencing and mitochondrial proteomics with assessments of metabolic and contractile function, we investigated the roles of dystrophin and utrophin in fast-to-slow muscle remodeling with low-frequency electrical stimulation (LFS, 10 Hz, 12 h/d, 7 d/wk, 28 d) in mdx (dystrophin null) and dko (dystrophin/utrophin null) mice, two established preclinical models of DMD. Results: Novel biological roles in adaptation were demonstrated by impaired transcriptional activation of estrogen-related receptor alpha-responsive genes supporting oxidative phosphorylation in dystrophic muscles. Further, utrophin expression in dystrophic muscles was required for LFS-induced remodeling of mitochondrial respiratory chain complexes, enhanced fiber respiration, and conferred protection from eccentric contraction-mediated damage. Conclusions: These findings reveal novel roles for dystrophin and utrophin during LFS-induced metabolic remodeling of dystrophic muscle and highlight the therapeutic potential of LFS to ameliorate the dystrophic pathology and protect from contraction-induced injury with important implications for DMD and related muscle disorders.
- Published
- 2021
- Full Text
- View/download PDF
5. Dissecting the Roles of Mitochondrial Complex I Intermediate Assembly Complex Factors in the Biogenesis of Complex I
- Author
-
Luke E. Formosa, Linden Muellner-Wong, Boris Reljic, Alice J. Sharpe, Thomas D. Jackson, Traude H. Beilharz, Diana Stojanovski, Michael Lazarou, David A. Stroud, and Michael T. Ryan
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Mitochondrial complex I harbors 7 mitochondrial and 38 nuclear-encoded subunits. Its biogenesis requires the assembly and integration of distinct intermediate modules, mediated by numerous assembly factors. The mitochondrial complex I intermediate assembly (MCIA) complex, containing assembly factors NDUFAF1, ECSIT, ACAD9, and TMEM126B, is required for building the intermediate ND2-module. The role of the MCIA complex and the involvement of other proteins in the biogenesis of this module is unclear. Cell knockout studies reveal that while each MCIA component is critical for complex I assembly, a hierarchy of stability exists centered on ACAD9. We also identify TMEM186 and COA1 as bona fide components of the MCIA complex with loss of either resulting in MCIA complex defects and reduced complex I assembly. TMEM186 enriches with newly translated ND3, and COA1 enriches with ND2. Our findings provide new functional insights into the essential nature of the MCIA complex in complex I assembly. : Formosa et al. investigate the function of the MCIA complex in complex I assembly. They demonstrate the requirement of individual components for the formation of complex I intermediates and assembly of the final enzyme. Finally, they characterize the involvement of TMEM186 and COA1 in this process. Keywords: assembly factors, complex I, MCIA complex, mitochondria, NADH-ubiquinone dehydrogenase, oxidative phosphorylation
- Published
- 2020
- Full Text
- View/download PDF
6. VDAC2 enables BAX to mediate apoptosis and limit tumor development
- Author
-
Hui San Chin, Mark X. Li, Iris K. L. Tan, Robert L. Ninnis, Boris Reljic, Kristen Scicluna, Laura F. Dagley, Jarrod J. Sandow, Gemma L. Kelly, Andre L. Samson, Stephane Chappaz, Seong L. Khaw, Catherine Chang, Andrew Morokoff, Kerstin Brinkmann, Andrew Webb, Colin Hockings, Cathrine M. Hall, Andrew J. Kueh, Michael T. Ryan, Ruth M. Kluck, Philippe Bouillet, Marco J. Herold, Daniel H. D. Gray, David C. S. Huang, Mark F. van Delft, and Grant Dewson
- Subjects
Science - Abstract
BAX and BAK are pro-apoptotic proteins whose activity is essential for the action of many anti-cancer drugs and to suppress tumorigenesis. Here, the authors perform a genome-wide CRISPR/Cas9 screen and identify VDAC2 as a promoter of BAX-mediated apoptosis that is important for an efficient chemotherapeutic response and to suppress tumor formation.
- Published
- 2018
- Full Text
- View/download PDF
7. Helicobacter pylori VacA toxin/subunit p34: targeting of an anion channel to the inner mitochondrial membrane.
- Author
-
Grazyna Domańska, Christian Motz, Michael Meinecke, Anke Harsman, Panagiotis Papatheodorou, Boris Reljic, Elke A Dian-Lothrop, Antoine Galmiche, Oliver Kepp, Lars Becker, Kathrin Günnewig, Richard Wagner, and Joachim Rassow
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The vacuolating toxin VacA, released by Helicobacter pylori, is an important virulence factor in the pathogenesis of gastritis and gastroduodenal ulcers. VacA contains two subunits: The p58 subunit mediates entry into target cells, and the p34 subunit mediates targeting to mitochondria and is essential for toxicity. In this study we found that targeting to mitochondria is dependent on a unique signal sequence of 32 uncharged amino acid residues at the p34 N-terminus. Mitochondrial import of p34 is mediated by the import receptor Tom20 and the import channel of the outer membrane TOM complex, leading to insertion of p34 into the mitochondrial inner membrane. p34 assembles in homo-hexamers of extraordinary high stability. CD spectra of the purified protein indicate a content of >40% beta-strands, similar to pore-forming beta-barrel proteins. p34 forms an anion channel with a conductivity of about 12 pS in 1.5 M KCl buffer. Oligomerization and channel formation are independent both of the 32 uncharged N-terminal residues and of the p58 subunit of the toxin. The conductivity is efficiently blocked by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB), a reagent known to inhibit VacA-mediated apoptosis. We conclude that p34 essentially acts as a small pore-forming toxin, targeted to the mitochondrial inner membrane by a special hydrophobic N-terminal signal.
- Published
- 2010
- Full Text
- View/download PDF
8. Mitochondrial E3 ubiquitin ligase MARCHF5 controls BAK apoptotic activity independently of BH3-only proteins
- Author
-
Allan Shuai Huang, Hui San Chin, Boris Reljic, Tirta M. Djajawi, Iris K. L. Tan, Jia-Nan Gong, David A. Stroud, David C. S. Huang, Mark F. van Delft, and Grant Dewson
- Subjects
Cell Biology ,Molecular Biology - Published
- 2022
9. High-intensity training induces non-stoichiometric changes in the mitochondrial proteome of human skeletal muscle without reorganisation of respiratory chain content
- Author
-
Kevin Huynh, Tegan Stait, Cesare Granata, Melinda T. Coughlan, David Bishop, Natalie A. Mellett, H. Janssen, Jujiao Kuang, Nikeisha J Caruana, Nicholas A. Jamnick, Peter J. Meikle, David A. Stroud, Boris Reljic, Adrienne Laskowski, David R. Thorburn, Javier Botella, and Ann E. Frazier
- Subjects
Proteomics ,Adult ,Male ,Adolescent ,Proteome ,Bioenergetics ,Biopsy ,In silico ,Science ,education ,Respiratory chain ,General Physics and Astronomy ,Oxidative phosphorylation ,High-Intensity Interval Training ,Biology ,Article ,Oxidative Phosphorylation ,General Biochemistry, Genetics and Molecular Biology ,Electron Transport ,Young Adult ,Adenosine Triphosphate ,medicine ,Humans ,Transcriptomics ,Muscle, Skeletal ,Multidisciplinary ,High intensity ,Skeletal muscle ,Energy metabolism ,General Chemistry ,Adaptation, Physiological ,Mitochondrial proteome ,Healthy Volunteers ,Mitochondria ,Cell biology ,medicine.anatomical_structure ,Quality of Life ,Electron flow ,Fat metabolism - Abstract
Mitochondrial defects are implicated in multiple diseases and aging. Exercise training is an accessible, inexpensive therapeutic intervention that can improve mitochondrial bioenergetics and quality of life. By combining multiple omics techniques with biochemical and in silico normalisation, we removed the bias arising from the training-induced increase in mitochondrial content to unearth an intricate and previously undemonstrated network of differentially prioritised mitochondrial adaptations. We show that changes in hundreds of transcripts, proteins, and lipids are not stoichiometrically linked to the overall increase in mitochondrial content. Our findings suggest enhancing electron flow to oxidative phosphorylation (OXPHOS) is more important to improve ATP generation than increasing the abundance of the OXPHOS machinery, and do not support the hypothesis that training-induced supercomplex formation enhances mitochondrial bioenergetics. Our study provides an analytical approach allowing unbiased and in-depth investigations of training-induced mitochondrial adaptations, challenging our current understanding, and calling for careful reinterpretation of previous findings., Exercise training can be therapeutic but how mitochondria respond remains unclear. Here, the authors use multiple omics techniques to reveal a complex network of non-stoichiometric mitochondrial adaptations that are prioritized or deprioritised during different phases of exercise training.
- Published
- 2021
10. Intact TP-53 function is essential for sustaining durable responses to BH3-mimetic drugs in leukemias
- Author
-
Rachel Thijssen, Edward Chew, Sarah S. Gabriel, Brandon J. Aubrey, Ashish Bajel, Andrew W. Roberts, Andreas Strasser, Marie Schoumacher, David A. Stroud, Chris D. Riffkin, Claudia Bruedigam, Donia M Moujalled, Natasha S Anstee, Tirta Mario Djajawi, Veronique Litalien, Ruth M. Kluck, Lin Tai, Andrew H. Wei, Thomas David Morley, Zhen Xu, Giovanna Pomilio, Sarah T. Diepstraten, Sarah MacRaild, Axel Kallies, Christoffer Flensburg, Boris Reljic, Steven W. Lane, Maoshan Chen, Catherine Chang, Gemma L. Kelly, David C. S. Huang, Fiona C. Brown, Sébastien Banquet, Michael A. Dengler, Melissa X Shi, and Ian J. Majewski
- Subjects
0301 basic medicine ,Myeloid ,Chronic lymphocytic leukemia ,Apoptosis ,Mice, SCID ,Biochemistry ,Oxidative Phosphorylation ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Mice, Inbred NOD ,Mice, Knockout ,Sulfonamides ,Hematology ,Indolizines ,Myeloid leukemia ,Neoplasm Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,medicine.drug ,medicine.medical_specialty ,Programmed cell death ,Morpholines ,Immunology ,Azacitidine ,Antineoplastic Agents ,03 medical and health sciences ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Internal medicine ,medicine ,Animals ,Humans ,business.industry ,Venetoclax ,Interleukin-2 Receptor alpha Subunit ,Cell Biology ,Bridged Bicyclo Compounds, Heterocyclic ,Genes, p53 ,Isoquinolines ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Xenograft Model Antitumor Assays ,Peptide Fragments ,030104 developmental biology ,chemistry ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,CRISPR-Cas Systems ,Tumor Suppressor Protein p53 ,Apoptosis Regulatory Proteins ,business ,DNA Damage - Abstract
Selective targeting of BCL-2 with the BH3-mimetic venetoclax has been a transformative treatment for patients with various leukemias. TP-53 controls apoptosis upstream of where BCL-2 and its prosurvival relatives, such as MCL-1, act. Therefore, targeting these prosurvival proteins could trigger apoptosis across diverse blood cancers, irrespective of TP53 mutation status. Indeed, targeting BCL-2 has produced clinically relevant responses in blood cancers with aberrant TP-53. However, in our study, TP53-mutated or -deficient myeloid and lymphoid leukemias outcompeted isogenic controls with intact TP-53, unless sufficient concentrations of BH3-mimetics targeting BCL-2 or MCL-1 were applied. Strikingly, tumor cells with TP-53 dysfunction escaped and thrived over time if inhibition of BCL-2 or MCL-1 was sublethal, in part because of an increased threshold for BAX/BAK activation in these cells. Our study revealed the key role of TP-53 in shaping long-term responses to BH3-mimetic drugs and reconciled the disparate pattern of initial clinical response to venetoclax, followed by subsequent treatment failure among patients with TP53-mutant chronic lymphocytic leukemia or acute myeloid leukemia. In contrast to BH3-mimetics targeting just BCL-2 or MCL-1 at doses that are individually sublethal, a combined BH3-mimetic approach targeting both prosurvival proteins enhanced lethality and durably suppressed the leukemia burden, regardless of TP53 mutation status. Our findings highlight the importance of using sufficiently lethal treatment strategies to maximize outcomes of patients with TP53-mutant disease. In addition, our findings caution against use of sublethal BH3-mimetic drug regimens that may enhance the risk of disease progression driven by emergent TP53-mutant clones.
- Published
- 2021
11. Circulating BiP/Grp78 is a novel prognostic marker for sepsis‐mediated immune cell death
- Author
-
Irvin Jose, Ashley Mansell, Liana Mackiewicz, Boris Reljic, Richard S. Hotchkiss, Hamsa Puthalakath, Joseph Menassa, Christina Nedeva, Pierre Faou, Marc Pellegrini, and Marcel Doerflinger
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Programmed cell death ,medicine.medical_treatment ,Apoptosis ,Inflammation ,Biochemistry ,Cell Line ,Sepsis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Blood serum ,Immune system ,medicine ,Animals ,Humans ,Endoplasmic Reticulum Chaperone BiP ,Molecular Biology ,Heat-Shock Proteins ,Mice, Knockout ,Bcl-2-Like Protein 11 ,Cell Death ,business.industry ,Macrophages ,Cell Biology ,Immunotherapy ,Macrophage Activation ,Prognosis ,medicine.disease ,Survival Analysis ,RAW 264.7 Cells ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Unfolded protein response ,medicine.symptom ,business ,Biomarkers - Abstract
Sepsis remains to be a major contributor to mortality in ICUs, and immune suppression caused by immune cell apoptosis determines the overall patient survival. However, diagnosis of sepsis-induced lymphopenia remains problematic with no accurate prognostic techniques or biomarkers for cell death available. Developing reliable prognostic tools for sepsis-mediated cell death is not only important for identifying patients at increased risk of immune suppression but also to monitor treatment progress of currently trialed immunotherapy strategies. We have previously shown an important role for endoplasmic reticulum stress (ER stress) in inducing sepsis-mediated cell death and here report on the identification of a secreted form of the ER chaperone BiP (immunoglobulin binding protein) as a novel circulating prognostic biomarker for immune cell death and ER stress during sepsis. Using biochemical purification and mass spectrometry coupled with an established in vitro sepsis cell death assay, we identified BiP/Grp78 as a factor secreted by lipopolysaccharide-activated macrophages that is capable of inducing cell death in target cells. Quantitative ELISA analysis showed significantly elevated levels of circulating BiP in mice undergoing polymicrobial sepsis, which was absent in Bim-/- mice that are protected from sepsis-induced lymphopenia. Using blood serum from human sepsis patients, we could detect a significant difference in levels of secreted BiP in sepsis patients compared to nonseptic controls, suggesting that secreted circulating BiP could indeed be used as a prognostic marker that is directly correlative to immune cell death during sepsis.
- Published
- 2020
12. The Mitochondrial Acyl-carrier Protein Interaction Network Highlights Important Roles for LYRM Family Members in Complex I and Mitoribosome Assembly
- Author
-
Baobei Lyu, Boris Reljic, Linden Muellner-Wong, David A. Stroud, Alice J. Sharpe, Dylan McGann, Michael T. Ryan, Marris G. Dibley, Felix Kraus, and Luke E. Formosa
- Subjects
Ribosomal Proteins ,Protein family ,Biology ,Transfection ,Biochemistry ,Analytical Chemistry ,Protein–protein interaction ,Mitochondrial Proteins ,Mitochondrial Ribosomes ,03 medical and health sciences ,Protein structure ,Ribosomal protein ,Mitochondrial ribosome ,Humans ,Amino Acid Sequence ,Protein Interaction Maps ,Molecular Biology ,030304 developmental biology ,Mitochondrial ribosome assembly ,0303 health sciences ,Electron Transport Complex I ,Research ,030302 biochemistry & molecular biology ,Cell biology ,HEK293 Cells ,Mitochondrial respiratory chain ,Isotope Labeling ,Chaperone (protein) ,Mitochondrial Membranes ,biology.protein ,Apoptosis Regulatory Proteins - Abstract
NDUFAB1 is the mitochondrial acyl carrier protein (ACP) essential for cell viability. Through its pantetheine-4′-phosphate post-translational modification, NDUFAB1 interacts with members of the leucine-tyrosine-arginine motif (LYRM) protein family. Although several LYRM proteins have been described to participate in a variety of defined processes, the functions of others remain either partially or entirely unknown. We profiled the interaction network of NDUFAB1 to reveal associations with 9 known LYRM proteins as well as more than 20 other proteins involved in mitochondrial respiratory chain complex and mitochondrial ribosome assembly. Subsequent knockout and interaction network studies in human cells revealed the LYRM member AltMiD51 to be important for optimal assembly of the large mitoribosome subunit, consistent with recent structural studies. In addition, we used proteomics coupled with topographical heat-mapping to reveal that knockout of LYRM2 impairs assembly of the NADH-dehydrogenase module of complex I, leading to defects in cellular respiration. Together, this work adds to the catalogue of functions executed by LYRM family of proteins in building mitochondrial complexes and emphasizes the common and essential role of NDUFAB1 as a protagonist in mitochondrial metabolism.
- Published
- 2020
13. E3 ubiquitin ligase MARCHF5 controls BAK apoptotic activity independently of BH3-only proteins
- Author
-
Shuai A. Huang, Hui San Chin, Boris Reljic, Tirta M. Djajawi, Iris K.L. Tan, David A. Stroud, David C.S. Huang, Mark F. van Delft, and Grant Dewson
- Subjects
biological phenomena, cell phenomena, and immunity - Abstract
SUMMARYIntrinsic apoptosis is principally governed by the BCL-2 family of proteins, but some non-BCL-2 proteins are also critical to control this process. To identify novel apoptosis regulators, we performed a genome-wide CRISPR-Cas9 library screen, and it identified the mitochondrial E3 ubiquitin ligase MARCHF5/MITOL/RNF153 as an important regulator of BAK apoptotic function. Deleting MARCHF5 in diverse cell lines dependent on BAK conferred profound resistance to BH3-mimetic drugs. The loss of MARCHF5 or its E3 ubiquitin ligase activity surprisingly drove BAK to adopt an activated conformation, with resistance to BH3-mimetics afforded by the formation of inhibitory complexes with pro-survival proteins MCL-1 and BCL-XL. Importantly, these changes to BAK conformation and pro-survival association occurred independently of BH3-only proteins and influence on pro-survival proteins. This study identifies a new mechanism by which MARCHF5 regulates apoptotic cell death and provides new insight into how cancer cells respond to BH3-mimetic drugs. These data also highlight the emerging role of ubiquitin signalling in apoptosis that may be exploited therapeutically.
- Published
- 2022
14. Mitochondrial COA7 is a heme-binding protein involved in the early stages of complex IV assembly
- Author
-
Luke E. Formosa, Boris Reljic, Alice J. Sharpe, Megan J. Maher, Linden Muellner-Wong, David A. Stroud, Shadi Maghool, and Michael T. Ryan
- Subjects
chemistry.chemical_compound ,Protein structure ,Biochemistry ,biology ,chemistry ,Heme binding ,Mitochondrial intermembrane space ,biology.protein ,Cytochrome c oxidase ,Heme ,Histidine ,Cofactor ,Biogenesis - Abstract
Cytochrome c oxidase assembly factor 7 (COA7) is a metazoan-specific assembly factor, critical for the biogenesis of mitochondrial complex IV (cytochrome c oxidase). Although mutations in COA7 have been linked in patients to complex IV assembly defects and neurological conditions such as peripheral neuropathy, ataxia and leukoencephalopathy, the precise role COA7 plays in the biogenesis of complex IV is not known. Here we show that the absence of COA7 leads to arrest of the complex IV assembly pathway at the initial step where the COX1 module is built, which requires incorporation of copper and heme cofactors. In solution, purified COA7 binds heme with micromolar affinity, through axial ligation to the central iron atom by histidine and methionine residues. Surprisingly, the crystal structure of COA7, determined to 2.4 Å resolution, reveals a ‘banana-shaped’ molecule composed of five helix-turn-helix (α/α) repeats, tethered by disulfide bonds, with a structure entirely distinct from proteins with characterized heme binding activities. We therefore propose a role for COA7 in heme binding/chaperoning in the mitochondrial intermembrane space, this activity being crucial for and providing a missing link in complex IV biogenesis.Significance StatementAssembly factors play key roles in the biogenesis of many mitochondrial protein complexes regulating their stability, activity and incorporation of essential cofactors. COA7 is a metazoan-specific assembly factor, the absence or mutation of which in humans accompanies complex IV assembly defects and neurological conditions. Here we report the crystal structure of COA7 to 2.4 Å resolution, revealing a ‘banana-shaped’ molecule composed of five helix-turn-helix (α/α) repeats, tethered by disulfide bonds. Characterization of pathogenic variants reveals significantly lower stabilities, correlating with the associated disease outcomes. Fascinatingly, COA7 binds heme with micromolar affinity, despite the fact that the protein structure does not resemble previously characterized heme-binding proteins. This provides a possible missing link for heme handling in the mitochondrial intermembrane space.
- Published
- 2021
15. Optic atrophy-associated TMEM126A is an assembly factor for the ND4-module of mitochondrial complex I
- Author
-
Michael T. Ryan, Alice J. Sharpe, Boris Reljic, Luke E. Formosa, Linden Muellner-Wong, and David A. Stroud
- Subjects
0301 basic medicine ,Mitochondrial DNA ,Mitochondrial disease ,Protein subunit ,Quantitative proteomics ,Mitochondrion ,Biology ,medicine.disease_cause ,DNA, Mitochondrial ,03 medical and health sciences ,Atrophy ,0302 clinical medicine ,Genome editing ,medicine ,Humans ,Gene ,TMEM126B ,Mutation ,Multidisciplinary ,Electron Transport Complex I ,Membrane Proteins ,NADH Dehydrogenase ,Biological Sciences ,medicine.disease ,Cell biology ,Mitochondria ,Optic Atrophy ,030104 developmental biology ,HEK293 Cells ,Membrane protein ,Function (biology) ,030217 neurology & neurosurgery - Abstract
Mitochondrial disease is a debilitating condition with a diverse genetic aetiology. Here, we report that TMEM126A, a protein that is mutated in patients with autosomal recessive optic atrophy, participates directly in the assembly of mitochondrial complex I. Using a combination of genome editing, interaction studies and quantitative proteomics, we find that loss of TMEM126A results in an isolated complex I deficiency and that TMEM126A interacts with a number of complex I subunits and assembly factors. Pulse-labelling interaction studies reveal that TMEM126A associates with the newly synthesised mtDNA-encoded ND4 subunit of complex I. Our findings indicate that TMEM126A is involved in the assembly of the ND4 distal membrane module of complex I. Importantly, we clarify that the function of TMEM126A is distinct from its paralogue TMEM126B, which acts in assembly of the ND2-module of complex I, helping to explain the differences in disease aetiology observed between these two genes.
- Published
- 2021
16. Metabolic remodeling of dystrophic skeletal muscle reveals biological roles for dystrophin and utrophin in adaptation and plasticity
- Author
-
Ashleigh M. Philp, Gregory R. Steinberg, Stefan M. Gehrig, Paula M. Miotto, Jin D. Chung, Matthew J. Watt, Andrew Philp, Gordon S. Lynch, Justin P. Hardee, René Koopman, Karen J.B. Martins, Boris Reljic, Jen Trieu, James G. Ryall, Kristy Swiderski, Timur Naim, and David A. Stroud
- Subjects
Male ,0301 basic medicine ,musculoskeletal diseases ,lcsh:Internal medicine ,Utrophin ,Duchenne muscular dystrophy ,030209 endocrinology & metabolism ,Muscle disorder ,Brief Communication ,Muscle adaptation ,Dystrophin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Muscular dystrophy ,Muscle, Skeletal ,lcsh:RC31-1245 ,Molecular Biology ,biology ,Skeletal muscle ,Cell Biology ,medicine.disease ,musculoskeletal system ,Adaptation, Physiological ,Mitochondria ,Cell biology ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,030104 developmental biology ,Mitochondrial respiratory chain ,medicine.anatomical_structure ,Metabolic Engineering ,Mice, Inbred mdx ,biology.protein ,Oxidative metabolism ,Muscle Contraction - Abstract
Objectives Preferential damage to fast, glycolytic myofibers is common in many muscle-wasting diseases, including Duchenne muscular dystrophy (DMD). Promoting an oxidative phenotype could protect muscles from damage and ameliorate the dystrophic pathology with therapeutic relevance, but developing efficacious strategies requires understanding currently unknown biological roles for dystrophin and utrophin in dystrophic muscle adaptation and plasticity. Methods Combining whole transcriptome RNA sequencing and mitochondrial proteomics with assessments of metabolic and contractile function, we investigated the roles of dystrophin and utrophin in fast-to-slow muscle remodeling with low-frequency electrical stimulation (LFS, 10 Hz, 12 h/d, 7 d/wk, 28 d) in mdx (dystrophin null) and dko (dystrophin/utrophin null) mice, two established preclinical models of DMD. Results Novel biological roles in adaptation were demonstrated by impaired transcriptional activation of estrogen-related receptor alpha-responsive genes supporting oxidative phosphorylation in dystrophic muscles. Further, utrophin expression in dystrophic muscles was required for LFS-induced remodeling of mitochondrial respiratory chain complexes, enhanced fiber respiration, and conferred protection from eccentric contraction-mediated damage. Conclusions These findings reveal novel roles for dystrophin and utrophin during LFS-induced metabolic remodeling of dystrophic muscle and highlight the therapeutic potential of LFS to ameliorate the dystrophic pathology and protect from contraction-induced injury with important implications for DMD and related muscle disorders., Graphical abstract Image 1, Highlights • Transcriptional remodeling to chronic low-frequency electrical stimulation (LFS) is impaired in dystrophic muscles. • Loss of dystrophin and utrophin in dystrophic muscles disrupts remodeling of mitochondrial complexes I-III to chronic LFS. • Loss of dystrophin and utrophin in dystrophic muscles abrogates improvements in fiber respiration after chronic LFS. • Loss of dystrophin and utrophin in dystrophic muscles compromises protection from contraction-induced injury after chronic LFS.
- Published
- 2021
17. Training-induced bioenergetic improvement in human skeletal muscle is associated with non-stoichiometric changes in the mitochondrial proteome without reorganization of respiratory chain content
- Author
-
Ann E. Frazier, David A. Stroud, Nicholas A. Jamnick, David R. Thorburn, Adrienne Laskowski, Melinda T. Coughlan, Javier Botella, Nikeisha J. Caruana, Boris Reljic, Kevin Huynh, Tegan Stait, David Bishop, H. Janssen, Cesare Granata, Natalie A. Mellett, Peter J. Meikle, and Jujiao Kuang
- Subjects
medicine.anatomical_structure ,Bioenergetics ,In silico ,Respiratory chain ,medicine ,Skeletal muscle ,Electron flow ,Oxidative phosphorylation ,Biology ,Mitochondrial proteome ,Cell biology - Abstract
SUMMARYMitochondrial defects are implicated in multiple diseases and aging. Exercise training is an accessible and inexpensive therapeutic intervention improving mitochondrial bioenergetics and quality of life. By combining a multi-omics approach with biochemical and in silico normalization, we removed the bias arising from the training-induced increase in human skeletal muscle mitochondrial content to unearth an intricate and previously undemonstrated network of differentially prioritized mitochondrial adaptations. We show that changes in hundreds of transcripts, proteins, and lipids are not stoichiometrically linked to the increase in mitochondrial content. We demonstrate that enhancing electron flow to oxidative phosphorylation (OXPHOS) is more important to improve ATP generation than increasing the abundance of the OXPHOS machinery, and that training-induced supercomplex formation does not confer enhancements in mitochondrial bioenergetics. Our study provides a new analytical approach allowing unbiased and in-depth investigations of training-induced mitochondrial adaptations, challenging our current understanding and calling for careful reinterpretation of previous findings.
- Published
- 2021
18. Abnormalities of mitochondrial dynamics and bioenergetics in neuronal cells from CDKL5 deficiency disorder
- Author
-
John Christodoulou, Anita F. Quigley, David A. Stroud, Tegan Stait, Sean Massey, Nicole J Van Bergen, Mirella Dottori, David R. Thorburn, Boris Reljic, Molly Ellery, and Luke E. Formosa
- Subjects
0301 basic medicine ,Male ,Proteomics ,Adolescent ,Induced Pluripotent Stem Cells ,CDKL5 ,Rett syndrome ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Biology ,Mitochondrion ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Mitochondrial Dynamics ,MECP2 ,03 medical and health sciences ,0302 clinical medicine ,Neurodevelopmental disorder ,Cell Line, Tumor ,medicine ,Humans ,Oxidative phosphorylation ,Induced pluripotent stem cell ,Cells, Cultured ,Neurons ,Mutation ,Infant ,Cell Differentiation ,medicine.disease ,Cell biology ,Mitochondria ,CDKL5 deficiency disorder ,030104 developmental biology ,Mitochondrial respiratory chain ,Neurology ,Child, Preschool ,Female ,Energy Metabolism ,Epileptic Syndromes ,Spasms, Infantile ,030217 neurology & neurosurgery ,RC321-571 - Abstract
CDKL5 deficiency disorder (CDD) is a rare neurodevelopmental disorder caused by pathogenic variants in the Cyclin-dependent kinase-like 5 (CDKL5) gene, resulting in dysfunctional CDKL5 protein. It predominantly affects females and causes seizures in the first few months of life, ultimately resulting in severe intellectual disability. In the absence of targeted therapies, treatment is currently only symptomatic. CDKL5 is a serine/threonine kinase that is highly expressed in the brain, with a critical role in neuronal development. Evidence of mitochondrial dysfunction in CDD is gathering, but has not been studied extensively. We used human patient-derived induced pluripotent stem cells with a pathogenic truncating mutation (p.Arg59*) and CRISPR/Cas9 gene-corrected isogenic controls, differentiated into neurons, to investigate the impact of CDKL5 mutation on cellular function. Quantitative proteomics indicated mitochondrial defects in CDKL5 p.Arg59* neurons, and mitochondrial bioenergetics analysis confirmed decreased activity of mitochondrial respiratory chain complexes. Additionally, mitochondrial trafficking velocity was significantly impaired, and there was a higher percentage of stationary mitochondria. We propose mitochondrial dysfunction is contributing to CDD pathology, and should be a focus for development of targeted treatments for CDD.
- Published
- 2020
19. HIGD2A is Required for Assembly of the COX3 Module of Human Mitochondrial Complex IV
- Author
-
Ching-Seng Ang, Alison G. Compton, Michael T. Ryan, Hayley S. Mountford, Daniella H Hock, Linden Muellner-Wong, David A. Stroud, David R. Thorburn, and Boris Reljic
- Subjects
Respiratory chain ,Mitochondrion ,Biochemistry ,Mass Spectrometry ,Analytical Chemistry ,Electron Transport Complex IV ,Mitochondrial Proteins ,03 medical and health sciences ,Gene Knockout Techniques ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Chemistry ,Research ,030302 biochemistry & molecular biology ,HEK 293 cells ,Translation (biology) ,Cell biology ,Mitochondria ,Oxygen ,Mitochondrial respiratory chain ,HEK293 Cells ,Respirasome ,Mitochondrial Membranes ,Biogenesis - Abstract
Assembly factors play a critical role in the biogenesis of mitochondrial respiratory chain complexes I-IV where they assist in the membrane insertion of subunits, attachment of co-factors, and stabilization of assembly intermediates. The major fraction of complexes I, III and IV are present together in large molecular structures known as respiratory chain supercomplexes. Several assembly factors have been proposed as required for supercomplex assembly, including the hypoxia inducible gene 1 domain family member HIGD2A. Using gene-edited human cell lines and extensive steady state, translation and affinity enrichment proteomics techniques we show that loss of HIGD2A leads to defects in the de novo biogenesis of mtDNA-encoded COX3, subsequent accumulation of complex IV intermediates and turnover of COX3 partner proteins. Deletion of HIGD2A also leads to defective complex IV activity. The impact of HIGD2A loss on complex IV was not altered by growth under hypoxic conditions, consistent with its role being in basal complex IV assembly. Although in the absence of HIGD2A we show that mitochondria do contain an altered supercomplex assembly, we demonstrate it to harbor a crippled complex IV lacking COX3. Our results redefine HIGD2A as a classical assembly factor required for building the COX3 module of complex IV.
- Published
- 2020
20. Dissecting the Roles of Mitochondrial Complex I Intermediate Assembly Complex Factors in the Biogenesis of Complex I
- Author
-
Linden Muellner-Wong, David A. Stroud, Diana Stojanovski, Boris Reljic, Michael Lazarou, Luke E. Formosa, Thomas Daniel Jackson, Michael T. Ryan, Alice J. Sharpe, and Traude H. Beilharz
- Subjects
0301 basic medicine ,Electron Transport Complex I ,Organelle Biogenesis ,Chemistry ,Respiratory chain ,Computational biology ,Mitochondrion ,General Biochemistry, Genetics and Molecular Biology ,Oxidative Phosphorylation ,Mitochondria ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,lcsh:Biology (General) ,Humans ,Organelle biogenesis ,lcsh:QH301-705.5 ,030217 neurology & neurosurgery ,Mitochondrial Complex I ,Biogenesis ,TMEM126B - Abstract
Summary: Mitochondrial complex I harbors 7 mitochondrial and 38 nuclear-encoded subunits. Its biogenesis requires the assembly and integration of distinct intermediate modules, mediated by numerous assembly factors. The mitochondrial complex I intermediate assembly (MCIA) complex, containing assembly factors NDUFAF1, ECSIT, ACAD9, and TMEM126B, is required for building the intermediate ND2-module. The role of the MCIA complex and the involvement of other proteins in the biogenesis of this module is unclear. Cell knockout studies reveal that while each MCIA component is critical for complex I assembly, a hierarchy of stability exists centered on ACAD9. We also identify TMEM186 and COA1 as bona fide components of the MCIA complex with loss of either resulting in MCIA complex defects and reduced complex I assembly. TMEM186 enriches with newly translated ND3, and COA1 enriches with ND2. Our findings provide new functional insights into the essential nature of the MCIA complex in complex I assembly. : Formosa et al. investigate the function of the MCIA complex in complex I assembly. They demonstrate the requirement of individual components for the formation of complex I intermediates and assembly of the final enzyme. Finally, they characterize the involvement of TMEM186 and COA1 in this process. Keywords: assembly factors, complex I, MCIA complex, mitochondria, NADH-ubiquinone dehydrogenase, oxidative phosphorylation
- Published
- 2019
21. HIGD2A is required for assembly of the COX3 module of human mitochondrial complex IV
- Author
-
David A. Stroud, Hayley S. Mountford, Alison G. Compton, David R. Thorburn, Boris Reljic, Michael T. Ryan, Daniella H Hock, and Ching-Seng Ang
- Subjects
0303 health sciences ,Chemistry ,Respiratory chain ,Translation (biology) ,Mitochondrion ,Proteomics ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Mitochondrial respiratory chain ,Steady state (chemistry) ,030217 neurology & neurosurgery ,Biogenesis ,Domain family ,030304 developmental biology - Abstract
Assembly factors play a critical role in the biogenesis of mitochondrial respiratory chain complexes I-IV where they assist in the membrane insertion of subunits, attachment of co-factors, and stabilization of assembly intermediates. The major fraction of complexes I, III and IV are present together in large molecular structures known as respiratory chain supercomplexes. A number of assembly factors have been proposed as required for supercomplex assembly, including the hypoxia inducible gene 1 domain family member HIGD2A. Using gene-edited human cell lines and extensive steady state, translation and affinity enrichment proteomics techniques we show that loss of HIGD2A leads to defects in the de novo biogenesis of mtDNA-encoded COX3, subsequent accumulation of complex IV intermediates and turnover of COX3 partner proteins. Deletion of HIGD2A also leads to defective complex IV activity. The impact of HIGD2A loss on complex IV was not altered by growth under hypoxic conditions, consistent with its role being in basal complex IV assembly. While in the absence of HIGD2A we show that mitochondria do contain an altered supercomplex assembly, we demonstrate it to harbor a crippled complex IV lacking COX3. Our results redefine HIGD2A as a classical assembly factor required for building the COX3 module of complex IV.
- Published
- 2019
- Full Text
- View/download PDF
22. Distinct initiating events underpin the immune and metabolic heterogeneity of KRAS-mutant lung adenocarcinoma
- Author
-
Kate D. Sutherland, Sarah A. Best, Gavin M. Wright, Ariena Kersbergen, Sheryl Ding, James E Vince, Yi Xie, Kaiming Li, Ji-Ying Song, Vivek Rathi, Matthew E. Ritchie, Xueyi Dong, and Boris Reljic
- Subjects
Male ,0301 basic medicine ,Lung Neoplasms ,endocrine system diseases ,General Physics and Astronomy ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,lcsh:Science ,Kelch-Like ECH-Associated Protein 1 ,Multidisciplinary ,respiratory system ,Flow Cytometry ,Cancer metabolism ,Immunohistochemistry ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Female ,KRAS ,Cancer microenvironment ,NF-E2-Related Factor 2 ,Science ,Blotting, Western ,Adenocarcinoma of Lung ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Proto-Oncogene Proteins p21(ras) ,Alveolar cells ,03 medical and health sciences ,Immune system ,Cell Line, Tumor ,medicine ,Animals ,Cancer models ,Lung cancer ,neoplasms ,Transcription factor ,Cancer ,General Chemistry ,medicine.disease ,digestive system diseases ,NFE2L2 ,respiratory tract diseases ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer research ,lcsh:Q ,Reactive Oxygen Species ,Non-small-cell lung cancer - Abstract
The KRAS oncoprotein, a critical driver in 33% of lung adenocarcinoma (LUAD), has remained an elusive clinical target due to its perceived undruggable nature. The identification of dependencies borne through common co-occurring mutations are sought to more effectively target KRAS-mutant lung cancer. Approximately 20% of KRAS-mutant LUAD carry loss-of-function mutations in KEAP1, a negative regulator of the antioxidant response transcription factor NFE2L2/NRF2. We demonstrate that Keap1-deficient KrasG12D lung tumors arise from a bronchiolar cell-of-origin, lacking pro-tumorigenic macrophages observed in tumors originating from alveolar cells. Keap1 loss activates the pentose phosphate pathway, inhibition of which, using 6-AN, abrogated tumor growth. These studies highlight alternative therapeutic approaches to specifically target this unique subset of KRAS-mutant LUAD cancers., Lung adenocarcinomas frequently harbour KRAS mutations, of which a subset are characterized by co-mutation of KEAP1. Here the authors show, in mice, that KrasG12D mutant tumours are metabolically distinct, with a bronchiolar cell-of-origin.
- Published
- 2019
23. Mitochondrial peptide BRAWNIN is essential for vertebrate respiratory complex III assembly
- Author
-
Bruno Reversade, Shan Zhang, Loo Chien Wang, Srikanth Nama, Lena Ho, Ruey-Kuang Cheng, Caroline Lei Wee, Volodimir Olexiouk, Lisa Tucker-Kellogg, Gio Fidelito, Paula Duek Roggli, Joel Celio Francisco, Claire Tang, Prabha Sampath, Baptiste Kerouanton, Suresh Jesuthasan, Gerben Menschaert, Chao Liang, David A. Stroud, Lei Sun, Lydie Lane, Jih Hou Peh, Enrico Petretto, Boris Reljic, Radoslaw M. Sobota, Narendra Suhas Jagannathan, and Camille Mary
- Subjects
0301 basic medicine ,Male ,Proteomics ,PROTEOMICS DATA ,Respiratory chain ,General Physics and Astronomy ,PROTEIN ,Mitochondrion ,MICROPEPTIDE ,Oxidative Phosphorylation ,Open Reading Frames/genetics ,Animals, Genetically Modified ,Electron Transport Complex III ,Models ,Electron Transport Complex III/metabolism ,lcsh:Science ,Zebrafish ,Growth Disorders ,ddc:616 ,Multidisciplinary ,biology ,Mitochondrial Proteins/genetics/metabolism ,SMALL ORFS ,Cell biology ,Mitochondria ,MICROPROTEIN ,Mechanisms of disease ,Lactic acidosis ,Zebrafish/genetics/growth & development ,Gene Knockdown Techniques ,Proteome ,Models, Animal ,Acidosis, Lactic ,Female ,Acidosis ,Peptides/genetics/metabolism ,DATABASE ,Science ,Genetically Modified ,Zebrafish Proteins/genetics/metabolism ,Genetics and Molecular Biology ,Oxidative phosphorylation ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mitochondrial Proteins ,03 medical and health sciences ,Open Reading Frames ,Lactic/genetics ,Mitochondria/metabolism ,medicine ,Animals ,Humans ,Metabolomics ,030102 biochemistry & molecular biology ,IDENTIFICATION ,Animal ,Biology and Life Sciences ,Growth Disorders/genetics ,General Chemistry ,Energy metabolism ,Zebrafish Proteins ,biology.organism_classification ,medicine.disease ,Biological ,GENE ,SIGNAL ,Disease Models, Animal ,030104 developmental biology ,Disease Models ,General Biochemistry ,UPDATE ,lcsh:Q ,Peptides ,Flux (metabolism) - Abstract
The emergence of small open reading frame (sORF)-encoded peptides (SEPs) is rapidly expanding the known proteome at the lower end of the size distribution. Here, we show that the mitochondrial proteome, particularly the respiratory chain, is enriched for small proteins. Using a prediction and validation pipeline for SEPs, we report the discovery of 16 endogenous nuclear encoded, mitochondrial-localized SEPs (mito-SEPs). Through functional prediction, proteomics, metabolomics and metabolic flux modeling, we demonstrate that BRAWNIN, a 71 a.a. peptide encoded by C12orf73, is essential for respiratory chain complex III (CIII) assembly. In human cells, BRAWNIN is induced by the energy-sensing AMPK pathway, and its depletion impairs mitochondrial ATP production. In zebrafish, Brawnin deletion causes complete CIII loss, resulting in severe growth retardation, lactic acidosis and early death. Our findings demonstrate that BRAWNIN is essential for vertebrate oxidative phosphorylation. We propose that mito-SEPs are an untapped resource for essential regulators of oxidative metabolism., Small open reading frame-encoded peptides (SEPs), shorter than 100 amino acids, are involved in many cell biological processes. Here the authors identify 16 nuclear-encoded mitochondrial SEPs, including BRAWNIN, an essential regulator of respiratory chain complex III assembly and ATP production.
- Published
- 2019
24. P62.05 Identifying Therapeutic Approaches to Treat KEAP1-Mutant Lung Adenocarcinoma
- Author
-
Matthew E. Ritchie, Ariena Kersbergen, Vivek Rathi, D. Desouza, Sheryl Ding, Malcolm J. McConville, Gavin M. Wright, Kate D. Sutherland, Boris Reljic, and Sarah A. Best
- Subjects
Pulmonary and Respiratory Medicine ,Lung ,medicine.anatomical_structure ,Oncology ,business.industry ,Mutant ,medicine ,Cancer research ,Adenocarcinoma ,medicine.disease ,business ,KEAP1 - Published
- 2021
25. Abstract PR03: Modeling the genetic heterogeneity of KRAS mutant lung adenocarcinomas for therapeutic discovery
- Author
-
Boris Reljic, Sarah A. Best, Ji-Ying Song, Sheryl Ding, and Kate D. Sutherland
- Subjects
Cancer Research ,Genetic heterogeneity ,Mutant ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,NFE2L2 ,Malignant transformation ,Oncology ,medicine ,Cancer research ,Adenocarcinoma ,KRAS ,Lung cancer ,Molecular Biology - Abstract
Lung adenocarcinoma (ADC) is the most common histologic subtype of lung cancer. Oncogenic driver mutations in KRAS are found in 36% of lung ADC cases and with a lack of specific KRAS inhibitors, this subset presents a significant clinical challenge. There is therefore an urgency to identify actionable co-occurring mutations as an alternative therapeutic approach. One of the most critical stress-response pathways in mammals is mediated by the transcription factor Nuclear Factor Erythroid-2-Related Factor 2 (NFE2L2/NRF2), which is negatively regulated by Kelch-like ECH-Associated Protein 1 (KEAP1). Loss-of-function mutations in KEAP1 have been identified in 19% of ADC, and often co-occur with KRAS mutations, but appear to be largely mutually exclusive with TP53 mutations. To best model these genetic subgroups of KRAS mutant lung ADC, we generated genetically engineered mouse models (GEMMs) whereby oncogenic KrasG12D was expressed either in combination with loss of p53 (KP mice) or Keap1 (KK mice). Critically, loss of Keap1 promoted KrasG12D-induced malignant transformation, which was associated with a unique metabolic program that could be exploited using pharmacologic inhibitors. Furthermore, bronchiolar epithelial cells appeared to be more sensitive than alveolar epithelial cells to transformation following Keap1 loss. Interestingly, this cell-type bias appeared to directly modulate the immune phenotype of lung tumors. Taken together, we have generated powerful in vivo models that enable us to address the relationship between tumor cells and resident immune cells to evaluate novel treatment strategies aimed at improving the long-term outcomes for patients carrying KRAS alterations. This abstract is also being presented as Poster B10. Citation Format: Sarah A. Best, Sheryl Ding, Boris Reljic, Ji-Ying Song, Kate D. Sutherland. Modeling the genetic heterogeneity of KRAS mutant lung adenocarcinomas for therapeutic discovery [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr PR03.
- Published
- 2020
26. VDAC2 enables BAX to mediate apoptosis and limit tumor development
- Author
-
Kristen Scicluna, Kerstin Brinkmann, Boris Reljic, Philippe Bouillet, Stephane Chappaz, Grant Dewson, Andrew P. Morokoff, Catherine Chang, Robert L Ninnis, Seong Lin Khaw, Laura F. Dagley, Ruth M. Kluck, Cathrine Hall, Andre L. Samson, Iris K. L. Tan, Gemma L. Kelly, Andrew I. Webb, Andrew J. Kueh, Marco J Herold, Daniel H.D. Gray, Hui San Chin, Michael T. Ryan, Mark F. van Delft, David C.S. Huang, Mark Xiang Li, Colin Hockings, and Jarrod J. Sandow
- Subjects
0301 basic medicine ,Programmed cell death ,Carcinogenesis ,Science ,Embryonic Development ,General Physics and Astronomy ,Apoptosis ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Bcl-2-associated X protein ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,lcsh:Science ,bcl-2-Associated X Protein ,Multidisciplinary ,biology ,Voltage-Dependent Anion Channel 2 ,Chemistry ,Intrinsic apoptosis ,General Chemistry ,HCT116 Cells ,Mitochondria ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,bcl-2 Homologous Antagonist-Killer Protein ,030104 developmental biology ,biology.protein ,lcsh:Q ,CRISPR-Cas Systems ,biological phenomena, cell phenomena, and immunity ,VDAC2 ,VDAC1 ,Bcl-2 Homologous Antagonist-Killer Protein ,HeLa Cells - Abstract
Intrinsic apoptosis is critical to prevent tumor formation and is engaged by many anti-cancer agents to eliminate tumor cells. BAX and BAK, the two essential mediators of apoptosis, are thought to be regulated through similar mechanisms and act redundantly to drive apoptotic cell death. From an unbiased genome-wide CRISPR/Cas9 screen, we identified VDAC2 (voltage-dependent anion channel 2) as important for BAX, but not BAK, to function. Genetic deletion of VDAC2 abrogated the association of BAX and BAK with mitochondrial complexes containing VDAC1, VDAC2, and VDAC3, but only inhibited BAX apoptotic function. Deleting VDAC2 phenocopied the loss of BAX in impairing both the killing of tumor cells by anti-cancer agents and the ability to suppress tumor formation. Together, our studies show that efficient BAX-mediated apoptosis depends on VDAC2, and reveal a striking difference in how BAX and BAK are functionally impacted by their interactions with VDAC2., BAX and BAK are pro-apoptotic proteins whose activity is essential for the action of many anti-cancer drugs and to suppress tumorigenesis. Here, the authors perform a genome-wide CRISPR/Cas9 screen and identify VDAC2 as a promoter of BAX-mediated apoptosis that is important for an efficient chemotherapeutic response and to suppress tumor formation.
- Published
- 2018
27. In the absence of apoptosis, myeloid cells arrest when deprived of growth factor, but remain viable by consuming extracellular glucose
- Author
-
David L. Vaux, Li Dong, Andrew G. Elefanty, Elizabeth Ng, Lisa M Lindqvist, Hoanh Tran, Boris Reljic, and Jen G. Cheung
- Subjects
0301 basic medicine ,Cell Survival ,medicine.medical_treatment ,Glucose uptake ,ATG5 ,Apoptosis ,Article ,Autophagy-Related Protein 5 ,Andrology ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,medicine ,Animals ,Myeloid Cells ,Molecular Biology ,030304 developmental biology ,Interleukin 3 ,bcl-2-Associated X Protein ,0303 health sciences ,biology ,Chemistry ,Growth factor ,fungi ,Glucose transporter ,Cell Biology ,030104 developmental biology ,Glucose ,bcl-2 Homologous Antagonist-Killer Protein ,030220 oncology & carcinogenesis ,biology.protein ,GLUT1 ,Interleukin-3 ,TXNIP ,Fetal bovine serum - Abstract
Withdrawal of the growth factor interleukin 3 from IL3-dependent myeloid cells causes them to undergo Bax/Bak1-dependent apoptosis, whereas factor-deprivedBax-/-Bak1-/-cells remain viable, but arrest and shrink. It was reported that withdrawal of IL3 fromBax-/-Bak1-/-cells caused decreased expression of the glucose transporter Glut1, leading to reduced glucose uptake, so that arrested cells required Atg5-dependent autophagy for long-term survival. In other cell types, a decrease in Glut1 is mediated by the thioredoxin-interacting protein Txnip, which is induced in IL3-dependent myeloid cells when growth factor is removed. We mutatedAtg5andTxnipby CRISPR/Cas9 and found that Atg5-dependent autophagy was not necessary for the long-term viability of cycling or arrestedBax-/-Bak1-/-cells, and that Txnip was not required for the decrease in Glut1 expression in response to IL3 withdrawal. Surprisingly, Atg5-deficientBax/Bak1double mutant cells survived for several weeks in medium supplemented with 10% fetal bovine serum (FBS), without high concentrations of added glucose or glutamine. When serum was withdrawn, the provision of an equivalent amount of glucose present in 10% FBS (~0.5 mM) was sufficient to support cell survival for more than a week, in the presence or absence of IL3. Thus,Bax-/-Bak1-/-myeloid cells deprived of growth factor consume extracellular glucose to maintain long-term viability, without a requirement for Atg5-dependent autophagy.
- Published
- 2018
28. BAX requires VDAC2 to mediate apoptosis and to limit tumor development
- Author
-
Stephane Chappaz, Michael T. Ryan, Colin Hockings, Andrew I. Webb, Boris Reljic, Kristen Scicluna, Seong Lin Khaw, Laura F. Dagley, Catherine Chang, Marco J Herold, Hui San Chin, Cathrine Hall, Grant Dewson, Ruth M. Kluck, Robert L Ninnis, Jarrod J. Sandow, Andrew J. Kueh, Daniel Hd Gray, David C.S. Huang, Mark F. van Delft, Gemma L. Kelly, Philippe Bouillet, Li X Mark, and Iris K. L. Tan
- Subjects
0303 health sciences ,VDAC3 ,Venetoclax ,Intrinsic apoptosis ,Mitochondrion ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Apoptosis ,Cytotoxic T cell ,biological phenomena, cell phenomena, and immunity ,VDAC2 ,VDAC1 ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Intrinsic apoptosis is critical for normal physiology including the prevention of tumor formation. BAX and BAK are essential for mediating this process and for the cytotoxic action of many anticancer drugs. BAX and BAK are thought to act in a functionally redundant manner and are considered to be regulated similarly. From an unbiased genome-wide CRISPR/Cas9 screen, we identified VDAC2 (voltage-dependent anion channel 2) as essential for BAX, but not BAK, to function. The genetic deletion of VDAC2 abrogated the association of BAX and BAK with mitochondrial complexes that contain VDAC1, VDAC2 and VDAC3. By disrupting its localization to mitochondria, BAX is rendered completely ineffective. Moreover, we defined an interface unique to VDAC2 that is required to drive BAX activity. Consequently, interfering with this interaction or deleting VDAC2 phenocopied the loss of BAX, including impairing the killing of tumor cells by anti-cancer agents such as the BCL-2 inhibitor venetoclax. Furthermore, the ability of BAX to prevent tumor formation was attenuated in the absence of VDAC2. Taken together, our studies show for the first time that BAX-mediated apoptosis, but not BAK-mediated apoptosis, is critically dependent on VDAC2, hence revealing the differential regulation of BAX and BAK.
- Published
- 2018
29. BAX-BAK1-independent LC3B lipidation by BH3 mimetics is unrelated to BH3 mimetic activity and has only minimal effects on autophagic flux
- Author
-
Erinna F. Lee, Boris Reljic, Jean-Marc Garnier, Guillaume Lessene, Lisa M Lindqvist, W. Douglas Fairlie, David L. Vaux, Stephanie A. Conos, Li Dong, Reljic, Boris, Conos, Stephanie, Lee, Erinna F, Garnier, Jean-Marc, Dong, Li, Lessene, Guillaume, Fairlie, W Douglas, Vaux, David L, and Lindqvist, Lisa M
- Subjects
0301 basic medicine ,autophagy ,BCL2 ,Programmed cell death ,Lipid-anchored protein ,Apoptosis ,Biology ,Piperazines ,Nitrophenols ,03 medical and health sciences ,Mice ,Bcl-2-associated X protein ,ABT-263 ,Autophagy ,Animals ,Molecular Biology ,Cells, Cultured ,bcl-2-Associated X Protein ,Sulfonamides ,Aniline Compounds ,ABT-737 ,fungi ,Biphenyl Compounds ,Basic Brief Report ,apoptosis ,BECN1 ,Cell Biology ,Bridged Bicyclo Compounds, Heterocyclic ,Cell biology ,Biphenyl compound ,cell death ,030104 developmental biology ,bcl-2 Homologous Antagonist-Killer Protein ,Proto-Oncogene Proteins c-bcl-2 ,ABT-199 ,biology.protein ,biological phenomena, cell phenomena, and immunity ,MAP1LC3B ,Microtubule-Associated Proteins ,Bcl-2 Homologous Antagonist-Killer Protein - Abstract
Inhibition of prosurvival BCL2 family members can induce autophagy, but the mechanism is controversial. We have provided genetic evidence that BCL2 family members block autophagy by inhibiting BAX and BAK1, but others have proposed they instead inhibit BECN1. Here we confirm that small molecule BH3 mimetics can induce BAX- and BAK1-independent MAP1LC3B/LC3B lipidation, but this only occurred at concentrations far greater than required to induce apoptosis and dissociate canonical BH3 domain-containing proteins that bind more tightly than BECN1. Because high concentrations of a less-active enantiomer of ABT-263 also induced BAX- and BAK1-independent LC3B lipidation, induction of this marker of autophagy appears to be an off-target effect. Indeed, robust autophagic flux was not induced by BH3 mimetic compounds in the absence of BAX and BAK1. Therefore at concentrations that are on target and achievable in vivo, BH3 mimetics only induce autophagy in a BAX- and BAK1-dependent manner. Refereed/Peer-reviewed
- Published
- 2016
30. Screening Strategies for TALEN-Mediated Gene Disruption
- Author
-
Boris Reljic and David A. Stroud
- Subjects
0301 basic medicine ,Genetics ,03 medical and health sciences ,Transcription activator-like effector nuclease ,030104 developmental biology ,Genome editing ,Cas9 ,Effector ,RNA splicing ,Gene targeting ,CRISPR ,Biology ,Gene - Abstract
Targeted gene disruption has rapidly become the tool of choice for the analysis of gene and protein function in routinely cultured mammalian cells. Three main technologies capable of irreversibly disrupting gene-expression exist: zinc-finger nucleases, transcription activator-like effector nucleases (TALENs), and the CRISPR/Cas9 system. The desired outcome of the use of any of these technologies is targeted insertions and/or deletions (indels) that result in either a nonsense frame shift or splicing error that disrupts protein expression. Many excellent do-it-yourself systems for TALEN construct assembly are now available at low or no cost to academic researchers. However, for new users, screening for successful gene disruption is still a hurdle. Here, we describe efficient and cost-effective strategies for the generation of gene-disrupted cell lines. Although the focus of this chapter is on the use of TALENs, these strategies can be applied to the use of all three technologies.
- Published
- 2016
31. MA24.10 Interrogating the Metabolic Effects of Keap1 Inactivation in Adenocarcinoma
- Author
-
Kate D. Sutherland, Sarah A. Best, Boris Reljic, Sheryl Ding, and Ariena Kersbergen
- Subjects
Pulmonary and Respiratory Medicine ,business.industry ,medicine.disease ,medicine.disease_cause ,KEAP1 ,Keap1 nrf2 ,Oncology ,Metabolic effects ,Cancer research ,Medicine ,Adenocarcinoma ,KRAS ,Respiratory system ,business - Published
- 2018
32. BAX-BAK1-independent LC3B lipidation by BH3 mimetics is unrelated to BH3 mimetic activity and has only minimal effects on autophagic flux
- Author
-
Boris Reljic, Stephanie Conos, Erinna F. Lee, Jean-Marc Garnier, Li Dong, Guillaume Lessene, W. Douglas Fairlie, David L. Vaux, Lisa M. Lindqvist, Boris Reljic, Stephanie Conos, Erinna F. Lee, Jean-Marc Garnier, Li Dong, Guillaume Lessene, W. Douglas Fairlie, David L. Vaux, and Lisa M. Lindqvist
- Abstract
Inhibition of prosurvival BCL2 family members can induce autophagy, but the mechanism is controversial. We have provided genetic evidence that BCL2 family members block autophagy by inhibiting BAX and BAK1, but others have proposed they instead inhibit BECN1. Here we confirm that small molecule BH3 mimetics can induce BAX- and BAK1-independent MAP1LC3B/LC3B lipidation, but this only occurred at concentrations far greater than required to induce apoptosis and dissociate canonical BH3 domain-containing proteins that bind more tightly than BECN1. Because high concentrations of a less-active enantiomer of ABT-263 also induced BAX- and BAK1-independent LC3B lipidation, induction of this marker of autophagy appears to be an off-target effect. Indeed, robust autophagic flux was not induced by BH3 mimetic compounds in the absence of BAX and BAK1. Therefore at concentrations that are on target and achievable in vivo, BH3 mimetics only induce autophagy in a BAX- and BAK1-dependent manner.
- Published
- 2016
- Full Text
- View/download PDF
33. Conversion of Bim-BH3 from Activator to Inhibitor of Bak through Structure-Based Design
- Author
-
Jason M. Brouwer, Ahmad Wardak, Grant Dewson, Mark F. van Delft, Adeline Y. Robin, Erinna F. Lee, Brad E. Sleebs, Iris K. L. Tan, Jonathan P. Bernardini, Peter M. Colman, W. Douglas Fairlie, Richard W Birkinshaw, Melissa J. Call, Brian J. Smith, Boris Reljic, Ping Lan, Guillaume Lessene, Peter E. Czabotar, and Angus D. Cowan
- Subjects
0301 basic medicine ,Programmed cell death ,Apoptosis ,Plasma protein binding ,Mitochondrion ,Biology ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Animals ,Humans ,Structure–activity relationship ,Molecular Biology ,Cell Line, Transformed ,Bcl-2-Like Protein 11 ,Activator (genetics) ,Bcl-2 family ,Cell Biology ,Mitochondria ,Cell biology ,bcl-2 Homologous Antagonist-Killer Protein ,030104 developmental biology ,biological phenomena, cell phenomena, and immunity ,Peptides ,Bacterial outer membrane ,Bcl-2 Homologous Antagonist-Killer Protein ,Protein Binding - Abstract
Certain BH3-only proteins transiently bind and activate Bak and Bax, initiating their oligomerization and the permeabilization of the mitochondrial outer membrane, a pivotal step in the mitochondrial pathway to apoptosis. Here we describe the first crystal structures of an activator BH3 peptide bound to Bak and illustrate their use in the design of BH3 derivatives capable of inhibiting human Bak on mitochondria. These BH3 derivatives compete for the activation site at the canonical groove, are the first engineered inhibitors of Bak activation, and support the role of key conformational transitions associated with Bak activation.
- Published
- 2017
34. Mitochondrial peptide BRAWNIN is essential for vertebrate respiratory complex III assembly
- Author
-
Shan Zhang, Boris Reljić, Chao Liang, Baptiste Kerouanton, Joel Celio Francisco, Jih Hou Peh, Camille Mary, Narendra Suhas Jagannathan, Volodimir Olexiouk, Claire Tang, Gio Fidelito, Srikanth Nama, Ruey-Kuang Cheng, Caroline Lei Wee, Loo Chien Wang, Paula Duek Roggli, Prabha Sampath, Lydie Lane, Enrico Petretto, Radoslaw M. Sobota, Suresh Jesuthasan, Lisa Tucker-Kellogg, Bruno Reversade, Gerben Menschaert, Lei Sun, David A. Stroud, and Lena Ho
- Subjects
Science - Abstract
Small open reading frame-encoded peptides (SEPs), shorter than 100 amino acids, are involved in many cell biological processes. Here the authors identify 16 nuclear-encoded mitochondrial SEPs, including BRAWNIN, an essential regulator of respiratory chain complex III assembly and ATP production.
- Published
- 2020
- Full Text
- View/download PDF
35. Mutations in the UQCC1-Interacting Protein, UQCC2, Cause Human Complex III Deficiency Associated with Perturbed Cytochrome b Protein Expression
- Author
-
John Christodoulou, Anne E. M. Leenders, Elena J. Tucker, Hayley S. Mountford, Hitoshi Endo, Alison G. Compton, Richard J. Rodenburg, Radek Szklarczyk, Damien L. Bruno, Ann E. Frazier, Leo G.J. Nijtmans, Martijn A. Huynen, Bas F.J. Wanschers, Boris Reljic, Michael T. Ryan, Mariël A.M. van den Brand, David R. Thorburn, Xiaonan W. Wijeyeratne, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Lab Centraal Lab (9), Complexe Genetica, and Genetica & Celbiologie
- Subjects
Cancer Research ,Small interfering RNA ,Mitochondrial Diseases ,Saccharomyces cerevisiae Proteins ,Cytochrome ,lcsh:QH426-470 ,Protein subunit ,Saccharomyces cerevisiae ,Mitochondrion ,Biology ,medicine.disease_cause ,Oxidative Phosphorylation ,Mitochondrial Proteins ,Genomic disorders and inherited multi-system disorders [IGMD 3] ,Consanguinity ,Electron Transport Complex III ,Renal tubular dysfunction ,Genetics ,medicine ,Humans ,Molecular Biology ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,Mutation ,Cytochrome b ,Homozygote ,Membrane Proteins ,Mitochondrial medicine Energy and redox metabolism [IGMD 8] ,Cytochromes b ,Fibroblasts ,Mitochondria ,lcsh:Genetics ,Mitochondrial medicine Membrane transport and intracellular motility [IGMD 8] ,Mitochondrial medicine [IGMD 8] ,Gene Expression Regulation ,Coenzyme Q – cytochrome c reductase ,biology.protein ,Energy and redox metabolism Mitochondrial medicine [NCMLS 4] ,Molecular Chaperones ,Research Article - Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) is responsible for generating the majority of cellular ATP. Complex III (ubiquinol-cytochrome c oxidoreductase) is the third of five OXPHOS complexes. Complex III assembly relies on the coordinated expression of the mitochondrial and nuclear genomes, with 10 subunits encoded by nuclear DNA and one by mitochondrial DNA (mtDNA). Complex III deficiency is a debilitating and often fatal disorder that can arise from mutations in complex III subunit genes or one of three known complex III assembly factors. The molecular cause for complex III deficiency in about half of cases, however, is unknown and there are likely many complex III assembly factors yet to be identified. Here, we used Massively Parallel Sequencing to identify a homozygous splicing mutation in the gene encoding Ubiquinol-Cytochrome c Reductase Complex Assembly Factor 2 (UQCC2) in a consanguineous Lebanese patient displaying complex III deficiency, severe intrauterine growth retardation, neonatal lactic acidosis and renal tubular dysfunction. We prove causality of the mutation via lentiviral correction studies in patient fibroblasts. Sequence-profile based orthology prediction shows UQCC2 is an ortholog of the Saccharomyces cerevisiae complex III assembly factor, Cbp6p, although its sequence has diverged substantially. Co-purification studies show that UQCC2 interacts with UQCC1, the predicted ortholog of the Cbp6p binding partner, Cbp3p. Fibroblasts from the patient with UQCC2 mutations have deficiency of UQCC1, while UQCC1-depleted cells have reduced levels of UQCC2 and complex III. We show that UQCC1 binds the newly synthesized mtDNA-encoded cytochrome b subunit of complex III and that UQCC2 patient fibroblasts have specific defects in the synthesis or stability of cytochrome b. This work reveals a new cause for complex III deficiency that can assist future patient diagnosis, and provides insight into human complex III assembly by establishing that UQCC1 and UQCC2 are complex III assembly factors participating in cytochrome b biogenesis., Author Summary Mitochondrial complex III deficiency is a devastating disorder that impairs energy generation, and leads to variable symptoms such as developmental regression, seizures, kidney dysfunction and frequently death. The genetic basis of complex III deficiency is not fully understood, with around half of cases having no known cause. This lack of genetic diagnosis is partly due to an incomplete understanding of the genes required for complex III assembly and function. We have identified two key proteins required for complex III, UQCC1 and UQCC2, and have elucidated the role of these inter-dependent proteins in the biogenesis of cytochrome b, the only complex III subunit that is encoded by mitochondrial DNA. We have shown that mutations in UQCC2 cause human complex III deficiency in a patient with neonatal lactic acidosis and renal tubulopathy. This work contributes to an improved understanding of complex III biogenesis, and will aid future molecular diagnoses of complex III deficiency.
- Published
- 2013
36. Abnormalities of mitochondrial dynamics and bioenergetics in neuronal cells from CDKL5 deficiency disorder
- Author
-
Nicole J. Van Bergen, Sean Massey, Tegan Stait, Molly Ellery, Boris Reljić, Luke E. Formosa, Anita Quigley, Mirella Dottori, David Thorburn, David A. Stroud, and John Christodoulou
- Subjects
CDKL5 ,CDKL5 deficiency disorder ,Mitochondria ,Induced pluripotent stem cell ,Oxidative phosphorylation ,Proteomics ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
CDKL5 deficiency disorder (CDD) is a rare neurodevelopmental disorder caused by pathogenic variants in the Cyclin-dependent kinase-like 5 (CDKL5) gene, resulting in dysfunctional CDKL5 protein. It predominantly affects females and causes seizures in the first few months of life, ultimately resulting in severe intellectual disability. In the absence of targeted therapies, treatment is currently only symptomatic. CDKL5 is a serine/threonine kinase that is highly expressed in the brain, with a critical role in neuronal development. Evidence of mitochondrial dysfunction in CDD is gathering, but has not been studied extensively. We used human patient-derived induced pluripotent stem cells with a pathogenic truncating mutation (p.Arg59*) and CRISPR/Cas9 gene-corrected isogenic controls, differentiated into neurons, to investigate the impact of CDKL5 mutation on cellular function. Quantitative proteomics indicated mitochondrial defects in CDKL5 p.Arg59* neurons, and mitochondrial bioenergetics analysis confirmed decreased activity of mitochondrial respiratory chain complexes. Additionally, mitochondrial trafficking velocity was significantly impaired, and there was a higher percentage of stationary mitochondria. We propose mitochondrial dysfunction is contributing to CDD pathology, and should be a focus for development of targeted treatments for CDD.
- Published
- 2021
- Full Text
- View/download PDF
37. Helicobacter pylori VacA Toxin/Subunit p34: Targeting of an Anion Channel to the Inner Mitochondrial Membrane
- Author
-
Elke A. Dian-Lothrop, Joachim Rassow, Antoine Galmiche, Oliver Kepp, Kathrin Günnewig, Grazyna Domanska, Anke Harsman, Christian Motz, Richard Wagner, Lars Becker, Michael Meinecke, Boris Reljic, and Panagiotis Papatheodorou
- Subjects
QH301-705.5 ,Protein subunit ,Immunology ,TIM/TOM complex ,Mitochondrion ,Biology ,Microbiology ,Mitochondrial apoptosis-induced channel ,Cell Biology/Membranes and Sorting ,Bacterial Proteins ,Virology ,Genetics ,Animals ,Humans ,Biology (General) ,Inner mitochondrial membrane ,Molecular Biology ,Helicobacter pylori ,RC581-607 ,Rats ,Electrophysiology ,Microscopy, Fluorescence ,Membrane protein ,Biochemistry ,Biophysics/Membrane Proteins and Energy Transduction ,Mitochondrial Membranes ,Translocase of the inner membrane ,Parasitology ,Immunologic diseases. Allergy ,Microbiology/Cellular Microbiology and Pathogenesis ,Bacterial outer membrane ,Research Article ,HeLa Cells - Abstract
The vacuolating toxin VacA, released by Helicobacter pylori, is an important virulence factor in the pathogenesis of gastritis and gastroduodenal ulcers. VacA contains two subunits: The p58 subunit mediates entry into target cells, and the p34 subunit mediates targeting to mitochondria and is essential for toxicity. In this study we found that targeting to mitochondria is dependent on a unique signal sequence of 32 uncharged amino acid residues at the p34 N-terminus. Mitochondrial import of p34 is mediated by the import receptor Tom20 and the import channel of the outer membrane TOM complex, leading to insertion of p34 into the mitochondrial inner membrane. p34 assembles in homo-hexamers of extraordinary high stability. CD spectra of the purified protein indicate a content of >40% β-strands, similar to pore-forming β-barrel proteins. p34 forms an anion channel with a conductivity of about 12 pS in 1.5 M KCl buffer. Oligomerization and channel formation are independent both of the 32 uncharged N-terminal residues and of the p58 subunit of the toxin. The conductivity is efficiently blocked by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB), a reagent known to inhibit VacA-mediated apoptosis. We conclude that p34 essentially acts as a small pore-forming toxin, targeted to the mitochondrial inner membrane by a special hydrophobic N-terminal signal., Author Summary VacA is a toxic protein produced by Helicobacter pylori, the bacteria that cause gastritis and ulcer diseases. p34, the toxic component of VacA, is known to damage mitochondria, defined cell organelles in the target cells. However, both the mechanism of mitochondrial targeting and the toxic activity inside the mitochondria are unclear. In this study, we show that p34 carries a unique targeting signal that is different from all targeting signatures that were previously identified in endogenous mitochondrial proteins. Eventually, p34 seems to act as an anion channel in the mitochondrial inner membrane and thus to destroy the balance of salt ions in the organelles.
- Published
- 2010
38. Mutations in the UQCC1-interacting protein, UQCC2, cause human complex III deficiency associated with perturbed cytochrome b protein expression.
- Author
-
Elena J Tucker, Bas F J Wanschers, Radek Szklarczyk, Hayley S Mountford, Xiaonan W Wijeyeratne, Mariël A M van den Brand, Anne M Leenders, Richard J Rodenburg, Boris Reljić, Alison G Compton, Ann E Frazier, Damien L Bruno, John Christodoulou, Hitoshi Endo, Michael T Ryan, Leo G Nijtmans, Martijn A Huynen, and David R Thorburn
- Subjects
Genetics ,QH426-470 - Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) is responsible for generating the majority of cellular ATP. Complex III (ubiquinol-cytochrome c oxidoreductase) is the third of five OXPHOS complexes. Complex III assembly relies on the coordinated expression of the mitochondrial and nuclear genomes, with 10 subunits encoded by nuclear DNA and one by mitochondrial DNA (mtDNA). Complex III deficiency is a debilitating and often fatal disorder that can arise from mutations in complex III subunit genes or one of three known complex III assembly factors. The molecular cause for complex III deficiency in about half of cases, however, is unknown and there are likely many complex III assembly factors yet to be identified. Here, we used Massively Parallel Sequencing to identify a homozygous splicing mutation in the gene encoding Ubiquinol-Cytochrome c Reductase Complex Assembly Factor 2 (UQCC2) in a consanguineous Lebanese patient displaying complex III deficiency, severe intrauterine growth retardation, neonatal lactic acidosis and renal tubular dysfunction. We prove causality of the mutation via lentiviral correction studies in patient fibroblasts. Sequence-profile based orthology prediction shows UQCC2 is an ortholog of the Saccharomyces cerevisiae complex III assembly factor, Cbp6p, although its sequence has diverged substantially. Co-purification studies show that UQCC2 interacts with UQCC1, the predicted ortholog of the Cbp6p binding partner, Cbp3p. Fibroblasts from the patient with UQCC2 mutations have deficiency of UQCC1, while UQCC1-depleted cells have reduced levels of UQCC2 and complex III. We show that UQCC1 binds the newly synthesized mtDNA-encoded cytochrome b subunit of complex III and that UQCC2 patient fibroblasts have specific defects in the synthesis or stability of cytochrome b. This work reveals a new cause for complex III deficiency that can assist future patient diagnosis, and provides insight into human complex III assembly by establishing that UQCC1 and UQCC2 are complex III assembly factors participating in cytochrome b biogenesis.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.