130 results on '"Gyorgy Szabadkai"'
Search Results
2. Macrophages induce malignant traits in mammary epithelium via IKKε/TBK1 kinases and the serine biosynthesis pathway
- Author
-
Ewa Wilcz‐Villega, Edward Carter, Alastair Ironside, Ruoyan Xu, Isabella Mataloni, Julie Holdsworth, William Jones, Rocío Moreno Béjar, Lukas Uhlik, Robert B Bentham, Susana A Godinho, Jesmond Dalli, Richard Grose, Gyorgy Szabadkai, Louise Jones, Kairbaan Hodivala‐Dilke, and Katiuscia Bianchi
- Subjects
inflammation ,macrophages ,malignant transformation ,obesity ,tumour metabolism ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract During obesity, macrophages infiltrate the breast tissue leading to low‐grade chronic inflammation, a factor considered responsible for the higher risk of breast cancer associated with obesity. Here, we formally demonstrate that breast epithelial cells acquire malignant properties when exposed to medium conditioned by macrophages derived from human healthy donors. These effects were mediated by the breast cancer oncogene IKKε and its downstream target—the serine biosynthesis pathway as demonstrated by genetic or pharmacological tools. Furthermore, amlexanox, an FDA‐approved drug targeting IKKε and its homologue TBK1, delayed in vivo tumour formation in a combined genetic mouse model of breast cancer and high‐fat diet‐induced obesity/inflammation. Finally, in human breast cancer tissues, we validated the link between inflammation–IKKε and alteration of cellular metabolism. Altogether, we identified a pathway connecting obesity‐driven inflammation to breast cancer and a potential therapeutic strategy to reduce the risk of breast cancer associated with obesity.
- Published
- 2020
- Full Text
- View/download PDF
3. Corrigendum: VHL-Mediated Regulation of CHCHD4 and Mitochondrial Function
- Author
-
Thomas Briston, Jenna M. Stephen, Luke W. Thomas, Cinzia Esposito, Yuen-Li Chung, Saiful E. Syafruddin, Mark Turmaine, Lucas A. Maddalena, Basma Greef, Gyorgy Szabadkai, Patrick H. Maxwell, Sakari Vanharanta, and Margaret Ashcroft
- Subjects
Hippel-Lindau protein (pVHL) ,hypoxia inducible factor ,mitochondria ,bioenergetics ,metabolism ,CHCHD4 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. CHCHD4 regulates tumour proliferation and EMT-related phenotypes, through respiratory chain-mediated metabolism
- Author
-
Luke W. Thomas, Cinzia Esposito, Jenna M. Stephen, Ana S. H. Costa, Christian Frezza, Thomas S. Blacker, Gyorgy Szabadkai, and Margaret Ashcroft
- Subjects
Coiled-coil helix coiled-coil helix domain-containing protein 4 (CHCHD4) ,hypoxia ,HIF-1α ,mitochondria ,respiratory chain ,disulfide relay system ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Mitochondrial oxidative phosphorylation (OXPHOS) via the respiratory chain is required for the maintenance of tumour cell proliferation and regulation of epithelial to mesenchymal transition (EMT)-related phenotypes through mechanisms that are not fully understood. The essential mitochondrial import protein coiled-coil helix coiled-coil helix domain-containing protein 4 (CHCHD4) controls respiratory chain complex activity and oxygen consumption, and regulates the growth of tumours in vivo. In this study, we interrogate the importance of CHCHD4-regulated mitochondrial metabolism for tumour cell proliferation and EMT-related phenotypes, and elucidate key pathways involved. Results Using in silico analyses of 967 tumour cell lines, and tumours from different cancer patient cohorts, we show that CHCHD4 expression positively correlates with OXPHOS and proliferative pathways including the mTORC1 signalling pathway. We show that CHCHD4 expression significantly correlates with the doubling time of a range of tumour cell lines, and that CHCHD4-mediated tumour cell growth and mTORC1 signalling is coupled to respiratory chain complex I (CI) activity. Using global metabolomics analysis, we show that CHCHD4 regulates amino acid metabolism, and that CHCHD4-mediated tumour cell growth is dependent on glutamine. We show that CHCHD4-mediated tumour cell growth is linked to CI-regulated mTORC1 signalling and amino acid metabolism. Finally, we show that CHCHD4 expression in tumours is inversely correlated with EMT-related gene expression, and that increased CHCHD4 expression in tumour cells modulates EMT-related phenotypes. Conclusions CHCHD4 drives tumour cell growth and activates mTORC1 signalling through its control of respiratory chain mediated metabolism and complex I biology, and also regulates EMT-related phenotypes of tumour cells.
- Published
- 2019
- Full Text
- View/download PDF
5. Vps34 PI 3-kinase inactivation enhances insulin sensitivity through reprogramming of mitochondrial metabolism
- Author
-
Benoit Bilanges, Samira Alliouachene, Wayne Pearce, Daniele Morelli, Gyorgy Szabadkai, Yuen-Li Chung, Gaëtan Chicanne, Colin Valet, Julia M. Hill, Peter J. Voshol, Lucy Collinson, Christopher Peddie, Khaled Ali, Essam Ghazaly, Vinothini Rajeeve, Georgios Trichas, Shankar Srinivas, Claire Chaussade, Rachel S. Salamon, Jonathan M. Backer, Cheryl L. Scudamore, Maria A. Whitehead, Erin P. Keaney, Leon O. Murphy, Robert K. Semple, Bernard Payrastre, Sharon A. Tooze, and Bart Vanhaesebroeck
- Subjects
Science - Abstract
Vps34 is a lipid kinase conserved from yeast to humans and involved in in intracellular vesicular trafficking and autophagy. Here Bilanges et al. show that inhibition of this kinase in mice improves glucose tolerance and diet-induced steatosis by modulating mitochondrial respiration and metabolism.
- Published
- 2017
- Full Text
- View/download PDF
6. Annual Meeting of the International Society of Cancer Metabolism (ISCaM): Metabolic Adaptations and Targets in Cancer
- Author
-
Sofia Avnet, Nicola Baldini, Lucie Brisson, Stine Falsig Pedersen, Paolo E. Porporato, Pierre Sonveaux, Gyorgy Szabadkai, and Silvia Pastorekova
- Subjects
cancer metabolism ,proton dynamics ,tumor microenvironment ,cancer imaging ,cancer therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The metabolism of cancer cells differs from that of their normal counterparts in a spectrum of attributes, including imbalances in diverse metabolic arms and pathways, metabolic plasticity and extent of adaptive responses, levels, and activities of metabolic enzymes and their upstream regulators and abnormal fluxes of metabolic intermediates and products. These attributes endow cancer cells with the ability to survive stressors of the tumor microenvironment and enable them to landscape and exploit the host terrain, thereby facilitating cancer progression and therapy resistance. Understanding the molecular and physiological principles of cancer metabolism is one of the key prerequisites for the development of better anticancer treatments. Therefore, various aspects of cancer metabolism were addressed at the 5th annual meeting of the International Society of Cancer Metabolism (ISCaM) in Bratislava, Slovakia, on October 17–20, 2018. The meeting presentations and discussions were traditionally focused on mechanistic, translational, and clinical characteristics of metabolism and pH control in cancer, at the level of molecular pathways, cells, tissues, and organisms. In order to reflect major healthcare challenges of the current era, ISCaM has extended its scope to metabolic disorders contributing to cancer, as well as to opportunities for their prevention, intervention, and therapeutic targeting.
- Published
- 2019
- Full Text
- View/download PDF
7. The mitochondrial calcium uniporter regulates breast cancer progression via HIF‐1α
- Author
-
Anna Tosatto, Roberta Sommaggio, Carsten Kummerow, Robert B Bentham, Thomas S Blacker, Tunde Berecz, Michael R Duchen, Antonio Rosato, Ivan Bogeski, Gyorgy Szabadkai, Rosario Rizzuto, and Cristina Mammucari
- Subjects
breast cancer ,HIF‐1α ,metastasis ,mitochondrial Ca2+ uptake ,reactive oxygen species ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Triple‐negative breast cancer (TNBC) represents the most aggressive breast tumor subtype. However, the molecular determinants responsible for the metastatic TNBC phenotype are only partially understood. We here show that expression of the mitochondrial calcium uniporter (MCU), the selective channel responsible for mitochondrial Ca2+ uptake, correlates with tumor size and lymph node infiltration, suggesting that mitochondrial Ca2+ uptake might be instrumental for tumor growth and metastatic formation. Accordingly, MCU downregulation hampered cell motility and invasiveness and reduced tumor growth, lymph node infiltration, and lung metastasis in TNBC xenografts. In MCU‐silenced cells, production of mitochondrial reactive oxygen species (mROS) is blunted and expression of the hypoxia‐inducible factor‐1α (HIF‐1α) is reduced, suggesting a signaling role for mROS and HIF‐1α, downstream of mitochondrial Ca2+. Finally, in breast cancer mRNA samples, a positive correlation of MCU expression with HIF‐1α signaling route is present. Our results indicate that MCU plays a central role in TNBC growth and metastasis formation and suggest that mitochondrial Ca2+ uptake is a potential novel therapeutic target for clinical intervention.
- Published
- 2016
- Full Text
- View/download PDF
8. VHL-Mediated Regulation of CHCHD4 and Mitochondrial Function
- Author
-
Thomas Briston, Jenna M. Stephen, Luke W. Thomas, Cinzia Esposito, Yuen-Li Chung, Saiful E. Syafruddin, Mark Turmaine, Lucas A. Maddalena, Basma Greef, Gyorgy Szabadkai, Patrick H. Maxwell, Sakari Vanharanta, and Margaret Ashcroft
- Subjects
von Hippel-Lindau protein (pVHL) ,hypoxia inducible factor ,mitochondria ,bioenergetics ,metabolism ,CHCHD4 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Dysregulated mitochondrial function is associated with the pathology of a wide range of diseases including renal disease and cancer. Thus, investigating regulators of mitochondrial function is of particular interest. Previous work has shown that the von Hippel-Lindau tumor suppressor protein (pVHL) regulates mitochondrial biogenesis and respiratory chain function. pVHL is best known as an E3-ubiquitin ligase for the α-subunit of the hypoxia inducible factor (HIF) family of dimeric transcription factors. In normoxia, pVHL recognizes and binds hydroxylated HIF-α (HIF-1α and HIF-2α), targeting it for ubiquitination and proteasomal degradation. In this way, HIF transcriptional activity is tightly controlled at the level of HIF-α protein stability. At least 80% of clear cell renal carcinomas exhibit inactivation of the VHL gene, which leads to HIF-α protein stabilization and constitutive HIF activation. Constitutive HIF activation in renal carcinoma drives tumor progression and metastasis. Reconstitution of wild-type VHL protein (pVHL) in pVHL-defective renal carcinoma cells not only suppresses HIF activation and tumor growth, but also enhances mitochondrial respiratory chain function via mechanisms that are not fully elucidated. Here, we show that pVHL regulates mitochondrial function when re-expressed in pVHL-defective 786O and RCC10 renal carcinoma cells distinct from its regulation of HIF-α. Expression of CHCHD4, a key component of the disulphide relay system (DRS) involved in mitochondrial protein import within the intermembrane space (IMS) was elevated by pVHL re-expression alongside enhanced expression of respiratory chain subunits of complex I (NDUFB10) and complex IV (mtCO-2 and COX IV). These changes correlated with increased oxygen consumption rate (OCR) and dynamic changes in glucose and glutamine metabolism. Knockdown of HIF-2α also led to increased OCR, and elevated expression of CHCHD4, NDUFB10, and COXIV in 786O cells. Expression of pVHL mutant proteins (R200W, N78S, D126N, and S183L) that constitutively stabilize HIF-α but differentially promote glycolytic metabolism, were also found to differentially promote the pVHL-mediated mitochondrial phenotype. Parallel changes in mitochondrial morphology and the mitochondrial network were observed. Our study reveals a new role for pVHL in regulating CHCHD4 and mitochondrial function in renal carcinoma cells.
- Published
- 2018
- Full Text
- View/download PDF
9. Annual Meeting of the International Society of Cancer Metabolism (ISCaM): Cancer Metabolism
- Author
-
Sofia Avnet, Nicola Baldini, Lucie Brisson, Angelo De Milito, Angela M. Otto, Silvia Pastoreková, Paolo E. Porporato, Gyorgy Szabadkai, and Pierre Sonveaux
- Subjects
tumor metabolism ,proton dynamics ,tumor microenvironment ,cancer imaging ,cancer therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Tumors are metabolic entities wherein cancer cells adapt their metabolism to their oncogenic agenda and microenvironmental influences. Metabolically different cancer cell subpopulations collaborate to optimize nutrient delivery with respect to immediate bioenergetic and biosynthetic needs. They can also metabolically exploit host cells. These metabolic networks are directly linked with cancer progression, treatment, resistance, and relapse. Conversely, metabolic alterations in cancer are exploited for anticancer therapy, imaging, and stratification for personalized treatments. These topics were addressed at the 4th annual meeting of the International Society of Cancer Metabolism (ISCaM) in Bertinoro, Italy, on 19–21 October 2017.
- Published
- 2018
- Full Text
- View/download PDF
10. Assessment of Cellular Redox State Using NAD(P)H Fluorescence Intensity and Lifetime
- Author
-
Tom Blacker, Tunde Berecz, Michael Duchen, and Gyorgy Szabadkai
- Subjects
Biology (General) ,QH301-705.5 - Abstract
NADH and NADPH are redox cofactors, primarily involved in catabolic and anabolic metabolic processes respectively. In addition, NADPH plays an important role in cellular antioxidant defence. In live cells and tissues, the intensity of their spectrally-identical autofluorescence, termed NAD(P)H, can be used to probe the mitochondrial redox state, while their distinct enzyme-binding characteristics can be used to separate their relative contributions to the total NAD(P)H intensity using fluorescence lifetime imaging microscopy (FLIM). These protocols allow differences in metabolism to be detected between cell types and altered physiological and pathological states.
- Published
- 2017
- Full Text
- View/download PDF
11. Discovery of multi-state gene cluster switches determining the adaptive mitochondrial and metabolic landscape of breast cancer
- Author
-
Michela Menegollo, Robert B. Bentham, Tiago Henriques, Seow Qi Ng, Ziyu Ren, Clarinde Esculier, Sia Agarwal, Emily Tong, Clement Lo, Sanjana Ilangovan, Zorka Szabadkai, Matteo Suman, Neill Patani, Avinash Ghanate, Kevin Bryson, Robert C. Stein, Mariia Yuneva, and Gyorgy Szabadkai
- Abstract
SummaryAdaptive metabolic switches are proposed to underlie conversions between cellular states during normal development as well as in cancer evolution, where they represent important therapeutic targets. However, the full spectrum, characteristics and regulation of existing metabolic switches are unknown. We propose that metabolic switches can be recognised by locating large alternating gene expression patterns and associate them with specific metabolic states. We developed a method to identify interspersed genesets by massive correlated biclustering (MCbiclust) and to predict their metabolic wiring. Testing the method on major breast cancer transcriptome datasets we discovered a series of gene sets with switch-like behaviour, predicting mitochondrial content, activity and central carbon fluxes in tumours associated with different switch positions. The predictions were experimentally validated by bioenergetic profiling and metabolic flux analysis of13C-labelled substrates and were ultimately extended by geneset analysis to link metabolic alterations to cellular states, thus predicting tumour pathology, prognosis and chemosensitivity. The method is applicable to any large and heterogeneous transcriptome dataset to discover metabolic and associated pathophysiological states.
- Published
- 2023
12. Impaired cellular bioenergetics caused by GBA1 depletion sensitizes neurons to calcium overload
- Author
-
Dany P. Perocheau, Gyorgy Szabadkai, Giulia Massaro, Ruggero Ferrazza, Nicoletta Plotegher, Gauri Bhosale, Michael R. Duchen, Ahad A. Rahim, Graziano Guella, Federico Zambon, and Simon N. Waddington
- Subjects
Free Radicals ,Bioenergetics ,Physiology ,Glutamic Acid ,chemistry.chemical_element ,Calcium ,Article ,Pathogenesis ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,medicine ,Animals ,Homeostasis ,Molecular Biology ,030304 developmental biology ,Membrane Potential, Mitochondrial ,Mice, Knockout ,Neurons ,Membrane potential ,0303 health sciences ,Neurodegeneration ,Autophagy ,Glutamate receptor ,Brain ,Correction ,Cell Biology ,Lipid Metabolism ,medicine.disease ,Mitochondria ,Cell biology ,Adenosine Diphosphate ,Receptors, Glutamate ,chemistry ,Glucosylceramidase ,Energy Metabolism ,Glucocerebrosidase ,030217 neurology & neurosurgery ,Neurological disorders ,Neuroscience - Abstract
Heterozygous mutations of the lysosomal enzyme glucocerebrosidase (GBA1) represent the major genetic risk for Parkinson’s disease (PD), while homozygous GBA1 mutations cause Gaucher disease, a lysosomal storage disorder, which may involve severe neurodegeneration. We have previously demonstrated impaired autophagy and proteasomal degradation pathways and mitochondrial dysfunction in neurons from GBA1 knockout (gba1−/−) mice. We now show that stimulation with physiological glutamate concentrations causes pathological [Ca2+]c responses and delayed calcium deregulation, collapse of mitochondrial membrane potential and an irreversible fall in the ATP/ADP ratio. Mitochondrial Ca2+ uptake was reduced in gba1−/− cells as was expression of the mitochondrial calcium uniporter. The rate of free radical generation was increased in gba1−/− neurons. Behavior of gba1+/− neurons was similar to gba1−/− in terms of all variables, consistent with a contribution of these mechanisms to the pathogenesis of PD. These data signpost reduced bioenergetic capacity and [Ca2+]c dysregulation as mechanisms driving neurodegeneration.
- Published
- 2019
13. Identification and functional validation of FDA-approved positive and negative modulators of the mitochondrial calcium uniporter
- Author
-
Gino A Cortopassi, Agnese De Mario, Julia M. Hill, Denis Vecellio Reane, Anna Tosatto, Rosario Rizzuto, Gyorgy Szabadkai, Janos Kriston-Vizi, Cristina Mammucari, and Robin Ketteler
- Subjects
0301 basic medicine ,Muscle Fibers, Skeletal ,Apoptosis ,benzethonium ,Pharmacology ,mitochondrial Ca2+ uptake ,Muscle hypertrophy ,Mice ,0302 clinical medicine ,Cell Movement ,Amorolfine ,Homeostasis ,media_common ,Chemistry ,Mitochondria ,Pharmaceutical Preparations ,mitochondrial calcium uniporter ,uptake ,triple-negative breast cancer ,Female ,medicine.drug ,Drug ,FDA-approved drugs ,media_common.quotation_subject ,High-throughput screening ,Morpholines ,2+ ,amorolfine ,Breast Neoplasms ,Duloxetine Hydrochloride ,high-throughput screening ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,skeletal muscle hypertrophy ,Oxygen Consumption ,Cell Line, Tumor ,medicine ,Gene silencing ,Animals ,Humans ,Calcium Signaling ,Muscle, Skeletal ,Cell Proliferation ,Benzethonium ,Cell growth ,United States Food and Drug Administration ,Reproducibility of Results ,Hypertrophy ,United States ,High-Throughput Screening Assays ,030104 developmental biology ,MCU ,Cytoprotection ,mitochondrial Ca ,Calcium ,Calcium Channels ,Energy Metabolism ,Reactive Oxygen Species ,030217 neurology & neurosurgery - Abstract
Summary The mitochondrial calcium uniporter (MCU), the highly selective channel responsible for mitochondrial Ca2+ entry, plays important roles in physiology and pathology. However, only few pharmacological compounds directly and selectively modulate its activity. Here, we perform high-throughput screening on a US Food and Drug Administration (FDA)-approved drug library comprising 1,600 compounds to identify molecules modulating mitochondrial Ca2+ uptake. We find amorolfine and benzethonium to be positive and negative MCU modulators, respectively. In agreement with the positive effect of MCU in muscle trophism, amorolfine increases muscle size, and MCU silencing is sufficient to blunt amorolfine-induced hypertrophy. Conversely, in the triple-negative breast cancer cell line MDA-MB-231, benzethonium delays cell growth and migration in an MCU-dependent manner and protects from ceramide-induced apoptosis, in line with the role of mitochondrial Ca2+ uptake in cancer progression. Overall, we identify amorolfine and benzethonium as effective MCU-targeting drugs applicable to a wide array of experimental and disease conditions., Graphical abstract, Highlights • We screen an FDA-approved drug library for mitochondrial Ca2+ uptake modulators • Amorolfine and benzethonium modulate MCU activity • Amorolfine increases MCU-dependent mitochondrial metabolism and muscle size • Benzethonium decreases MCU-dependent cancer cell growth and migration, In search of mitochondrial Ca2+ uptake modulators, De Mario et al. identify amorolfine and benzethonium as positive hits of a 1,600 FDA drug library high-throughput screen. Amorolfine increases mitochondrial metabolism and muscle size in an MCU-dependent manner. Benzethonium negatively regulates MCU activity, mROS formation, and cancer cell growth and migration.
- Published
- 2021
14. PICALM rescues glutamatergic neurotransmission, behavioural function and survival in a Drosophila model of Abeta42 toxicity
- Author
-
Teresa Niccoli, Linda Partridge, Gyorgy Szabadkai, Ziyu Ren, Benjamin Aleyakpo, Yifan Yu, and Nathaniel S. Woodling
- Subjects
AcademicSubjects/SCI01140 ,Vesicular Glutamate Transport Proteins ,Nerve Tissue Proteins ,Stimulation ,Neurotransmission ,Biology ,Receptors, Ionotropic Glutamate ,Synaptic Transmission ,PICALM ,03 medical and health sciences ,Glutamatergic ,0302 clinical medicine ,Alzheimer Disease ,Postsynaptic potential ,Genetics ,Animals ,Drosophila Proteins ,Humans ,Excitatory Amino Acid Agents ,Molecular Biology ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Amyloid beta-Peptides ,Behavior, Animal ,CCAAT-Enhancer-Binding Protein-beta ,Glutamate receptor ,General Medicine ,Peptide Fragments ,Drosophila melanogaster ,Monomeric Clathrin Assembly Proteins ,Amphiphysin ,General Article ,Neuroscience ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Alzheimer’s disease (AD) is the most common form of dementia and the most prevalent neurodegenerative disease. Genome-wide association studies have linked PICALM to AD risk. PICALM has been implicated in Aβ42 production and turnover, but whether it plays a direct role in modulating Aβ42 toxicity remains unclear. We found that increased expression of the Drosophila PICALM orthologue lap could rescue Aβ42 toxicity in an adult-onset model of AD, without affecting Aβ42 level. Imbalances in the glutamatergic system, leading to excessive, toxic stimulation, have been associated with AD. We found that Aβ42 caused the accumulation of presynaptic vesicular glutamate transporter (VGlut) and increased spontaneous glutamate release. Increased lap expression reversed these phenotypes back to control levels, suggesting that lap may modulate glutamatergic transmission. We also found that lap modulated the localization of amphiphysin (Amph), the homologue of another AD risk factor BIN1, and that Amph itself modulated postsynaptic glutamate receptor (GluRII) localization. We propose a model where PICALM modulates glutamatergic transmission, together with BIN1, to ameliorate synaptic dysfunction and disease progression., Graphical Abstract Graphical Abstractlap and Amph in glutamate metabolism. Aβ affects the localization of both VGlut and GluRIIA, leading to increased glutamate signalling. Lap and Amph restore glutamate signalling, potentially by promoting presynaptic endocytosis of VGlut via lap and/or Rab5/EndoA, and postsynaptic retrival of GluRIIA via Amph.
- Published
- 2020
15. Macrophages induce malignant traits in mammary epithelium via IKKε/TBK1 kinases and the serine biosynthesis pathway
- Author
-
William Jones, Julie Holdsworth, Louise Jones, Edward P. Carter, Lukas Uhlik, Isabella Mataloni, Robert B Bentham, Gyorgy Szabadkai, Richard Grose, Katiuscia Bianchi, Alastair Ironside, Susana A. Godinho, Rocío Moreno Béjar, Ewa Wilcz-Villega, Jesmond Dalli, Ruoyan Xu, and Kairbaan Hodivala-Dilke
- Subjects
0301 basic medicine ,Medicine (General) ,malignant transformation ,obesity ,Immunology ,Aminopyridines ,Breast Neoplasms ,Inflammation ,Protein Serine-Threonine Kinases ,QH426-470 ,Article ,Malignant transformation ,Serine ,Mice ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Breast cancer ,TANK-binding kinase 1 ,Genetics ,medicine ,Animals ,Humans ,Mammary Glands, Human ,skin and connective tissue diseases ,Cancer ,Oncogene ,Kinase ,business.industry ,Epithelial Cells ,Articles ,medicine.disease ,I-kappa B Kinase ,macrophages ,3. Good health ,inflammation ,tumour metabolism ,Metabolism ,030104 developmental biology ,Culture Media, Conditioned ,Cancer research ,Molecular Medicine ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
During obesity, macrophages infiltrate the breast tissue leading to low‐grade chronic inflammation, a factor considered responsible for the higher risk of breast cancer associated with obesity. Here, we formally demonstrate that breast epithelial cells acquire malignant properties when exposed to medium conditioned by macrophages derived from human healthy donors. These effects were mediated by the breast cancer oncogene IKKε and its downstream target—the serine biosynthesis pathway as demonstrated by genetic or pharmacological tools. Furthermore, amlexanox, an FDA‐approved drug targeting IKKε and its homologue TBK1, delayed in vivo tumour formation in a combined genetic mouse model of breast cancer and high‐fat diet‐induced obesity/inflammation. Finally, in human breast cancer tissues, we validated the link between inflammation–IKKε and alteration of cellular metabolism. Altogether, we identified a pathway connecting obesity‐driven inflammation to breast cancer and a potential therapeutic strategy to reduce the risk of breast cancer associated with obesity., In this article a new pathway has been identified that links macrophage‐driven inflammation and cellular metabolism to the acquisition of malignant traits in breast epithelium providing new insight into the well‐established association between breast cancer and obesity and offering a new preventive strategy.
- Published
- 2020
16. The breast cancer oncogene IKKε coordinates mitochondrial function and serine metabolism
- Author
-
Yewei Wang, Ai Nagano, Busra Yaman, Claude Chelala, Ana S. H. Costa, Kevin Bryson, Robert B Bentham, Pedro R. Cutillas, Christian Frezza, Ewa Wilcz-Villega, Ruoyan Xu, Gyorgy Szabadkai, Sheila Olendo Barasa, Katiuscia Bianchi, William Jones, Vinothini Rajeeve, Frezza, Christian [0000-0002-3293-7397], and Apollo - University of Cambridge Repository
- Subjects
IKKε ,mitochondrial metabolism ,Breast Neoplasms ,IκB kinase ,Activating Transcription Factor 4 ,Biology ,environment and public health ,Biochemistry ,Article ,Serine ,03 medical and health sciences ,0302 clinical medicine ,breast cancer ,Downregulation and upregulation ,Genetics ,Humans ,ATF4 ,skin and connective tissue diseases ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Cancer ,0303 health sciences ,Oncogene ,Chemistry ,Kinase ,Phosphoproteomics ,Articles ,Oncogenes ,I-kappa B Kinase ,Mitochondria ,enzymes and coenzymes (carbohydrates) ,Metabolism ,serine biosynthesis ,Cancer research ,Cytokine secretion ,Female ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
IκB kinase ε (IKKε) is a key molecule at the crossroads of inflammation and cancer. Known to regulate cytokine secretion via NFκB and IRF3, the kinase is also a breast cancer oncogene, overexpressed in a variety of tumours. However, to what extent IKKε remodels cellular metabolism is currently unknown. Here, we used metabolic tracer analysis to show that IKKε orchestrates a complex metabolic reprogramming that affects mitochondrial metabolism and consequently serine biosynthesis independently of its canonical signalling role. We found that IKKε upregulates the serine biosynthesis pathway (SBP) indirectly, by limiting glucose‐derived pyruvate utilisation in the TCA cycle, inhibiting oxidative phosphorylation. Inhibition of mitochondrial function induces activating transcription factor 4 (ATF4), which in turn drives upregulation of the expression of SBP genes. Importantly, pharmacological reversal of the IKKε‐induced metabolic phenotype reduces proliferation of breast cancer cells. Finally, we show that in a highly proliferative set of ER negative, basal breast tumours, IKKε and PSAT1 are both overexpressed, corroborating the link between IKKε and the SBP in the clinical context., The IκB kinase IKKε inhibits mitochondrial metabolism by limiting pyruvate oxidation, in turn activating the serine biosynthesis pathway via ATF4 in a NFκB and IRF3‐independent manner.
- Published
- 2020
17. Macrophages induce malignant traits in mammary epithelium via IKK epsilon/TBK1 kinases and the serine biosynthesis pathway
- Author
-
Wilcz-Villega, E., Carter, E., Ironside, A., Ry, Xu, Mataloni, I., Holdsworth, J., Jones, W., Bejar, Rm, Uhlik, L., Bentham, Rb, Godinho, Sa, Dalli, J., Grose, R., Gyorgy Szabadkai, Jones, L., Hodivala-Dilke, K., and Bianchi, K.
- Subjects
inflammation ,macrophages ,malignant transformation ,obesity ,tumour metabolism - Published
- 2020
18. Mitochondrial permeability transition pore: sensitivity to opening and mechanistic dependence on substrate availability
- Author
-
Sian Lewis, James M. Staddon, Gyorgy Szabadkai, Michael R. Duchen, Ben Powney, Malcolm Roberts, and Thomas Briston
- Subjects
0301 basic medicine ,Cellular respiration ,Cell Respiration ,Succinic Acid ,lcsh:Medicine ,Mitochondrion ,Mitochondrial Membrane Transport Proteins ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Oxygen Consumption ,0302 clinical medicine ,Animals ,Inner mitochondrial membrane ,lcsh:Science ,Membrane Potential, Mitochondrial ,chemistry.chemical_classification ,Membrane potential ,Reactive oxygen species ,Electron Transport Complex I ,Multidisciplinary ,Mitochondrial Permeability Transition Pore ,MPTP ,lcsh:R ,Hydrogen Peroxide ,NAD ,Mitochondria ,Rats ,030104 developmental biology ,chemistry ,Biochemistry ,Mitochondrial permeability transition pore ,Coenzyme Q – cytochrome c reductase ,Biophysics ,Calcium ,Female ,lcsh:Q ,Energy Metabolism ,Reactive Oxygen Species ,Ion Channel Gating ,030217 neurology & neurosurgery - Abstract
Mitochondrial Ca2+ uptake has a key role in cellular Ca2+ homeostasis. Excessive matrix Ca2+ concentrations, especially when coincident with oxidative stress, precipitate opening of an inner mitochondrial membrane, high-conductance channel: the mitochondrial permeability transition pore (mPTP). mPTP opening has been implicated as a final cell death pathway in numerous diseases and therefore understanding conditions dictating mPTP opening is crucial for developing targeted therapies. Here, we have investigated the impact of mitochondrial metabolic state on the probability and consequences of mPTP opening. Isolated mitochondria were energised using NADH- or FADH2-linked substrates. The functional consequences of Ca2+-induced mPTP opening were assessed by Ca2+ retention capacity, using fluorescence-based analysis, and simultaneous measurements of mitochondrial Ca2+ handling, membrane potential, respiratory rate and production of reactive oxygen species (ROS). Succinate-induced, membrane potential-dependent reverse electron transfer sensitised mitochondria to mPTP opening. mPTP-induced depolarisation under succinate subsequently inhibited reverse electron transfer. Complex I-driven respiration was reduced after mPTP opening but sustained in the presence of complex II-linked substrates, consistent with inhibition of complex I-supported respiration by leakage of matrix NADH. Additionally, ROS generated at complex III did not sensitise mitochondria to mPTP opening. Thus, cellular metabolic fluxes and metabolic environment dictate mitochondrial functional response to Ca2+ overload.
- Published
- 2017
19. MCbiclust: a novel algorithm to discover large-scale functionally related gene sets from massive transcriptomics data collections
- Author
-
Robert B Bentham, Kevin Bryson, and Gyorgy Szabadkai
- Subjects
Datasets as Topic ,Biology ,Synthetic data ,Biclustering ,Transcriptome ,Databases ,03 medical and health sciences ,0302 clinical medicine ,Genetic ,Neoplasms ,Databases, Genetic ,Genes, Regulator ,Genetics ,Cluster Analysis ,Humans ,Gene Regulatory Networks ,Gene ,Genetic Association Studies ,030304 developmental biology ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,Scale (chemistry) ,Gene Expression Profiling ,Regulator ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Identification (information) ,Phenotype ,Genes ,Gene Expression Regulation ,Algorithms ,Organelle biogenesis ,Algorithm ,030217 neurology & neurosurgery ,Network analysis - Abstract
The potential to understand fundamental biological processes from gene expression data has grown parallel with the recent explosion of the size of data collections. However, to exploit this potential, novel analytical methods are required, capable of handling massive data matrices. We found current methods limited in the size of correlated gene sets they could discover within biologically heterogeneous data collections, hampering the identification of multi-gene controlled fundamental cellular processes such as energy metabolism, organelle biogenesis and stress responses. Here we describe a novel biclustering algorithm called Massively Correlated Biclustering (MCbiclust) that selects samples and genes from large datasets with maximal correlated gene expression, allowing regulation of complex pathway to be examined. The method has been evaluated using synthetic data and applied to large bacterial and cancer cell datasets. We show that the large biclusters discovered, so far elusive to identification by existing techniques, are biologically relevant and thus MCbiclust has great potential use in the analysis of transcriptomics data to identify large scale unknown effects hidden within the data. The identified massive biclusters can be used to develop improved transcriptomics based diagnosis tools for diseases caused by altered gene expression, or used for further network analysis to understand genotype-phenotype correlations.
- Published
- 2017
20. MLH1 deficiency leads to deregulated mitochondrial metabolism
- Author
-
Claude Chelala, Danilo Cucchi, Sarah A. Martin, Stuart McDonald, Jun Wang, Marc J Williams, Andrew Silver, Marta O. Freitas, Gemma Bridge, Nirosha Suraweera, Sukaina Rashid, Gyorgy Szabadkai, and Zhi Yao
- Subjects
Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Mitochondrial DNA ,Immunology ,PINK1 ,Synthetic lethality ,Biology ,Transfection ,MLH1 ,DNA Mismatch Repair ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Neoplasms ,Rotenone ,Humans ,lcsh:QH573-671 ,Ovarian Neoplasms ,chemistry.chemical_classification ,Reactive oxygen species ,Electron Transport Complex I ,lcsh:Cytology ,DNA replication ,Cell Biology ,Metabolism ,HCT116 Cells ,Cancer metabolism ,digestive system diseases ,Endometrial Neoplasms ,Mitochondria ,Cell biology ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Mutation ,Female ,DNA mismatch repair ,Colorectal Neoplasms ,MutL Protein Homolog 1 ,HT29 Cells - Abstract
The DNA mismatch repair (MMR) pathway is responsible for the repair of base–base mismatches and insertion/deletion loops that arise during DNA replication. MMR deficiency is currently estimated to be present in 15–17% of colorectal cancer cases and 30% of endometrial cancers. MLH1 is one of the key proteins involved in the MMR pathway. Inhibition of a number of mitochondrial genes, including POLG and PINK1 can induce synthetic lethality in MLH1-deficient cells. Here we demonstrate for the first time that loss of MLH1 is associated with a deregulated mitochondrial metabolism, with reduced basal oxygen consumption rate and reduced spare respiratory capacity. Furthermore, MLH1-deficient cells display a significant reduction in activity of the respiratory chain Complex I. As a functional consequence of this perturbed mitochondrial metabolism, MLH1-deficient cells have a reduced anti-oxidant response and show increased sensitivity to reactive oxidative species (ROS)-inducing drugs. Taken together, our results provide evidence for an intrinsic mitochondrial dysfunction in MLH1-deficient cells and a requirement for MLH1 in the regulation of mitochondrial function.
- Published
- 2019
21. Metabolic Profiling of Live Cancer Tissues Using NAD(P)H Fluorescence Lifetime Imaging
- Author
-
Thomas S, Blacker, Michael D E, Sewell, Gyorgy, Szabadkai, and Michael R, Duchen
- Subjects
Data Analysis ,Microscopy, Fluorescence ,Research Design ,Liver Neoplasms ,Optical Imaging ,Metabolome ,Humans ,Metabolomics ,Energy Metabolism ,NADP - Abstract
Altered metabolism is a hallmark of cancer, both resulting from and driving oncogenesis. The NAD and NADP redox couples play a key role in a large number of the metabolic pathways involved. In their reduced forms, NADH and NADPH, these molecules are intrinsically fluorescent. As the average time for fluorescence to be emitted following excitation by a laser pulse, the fluorescence lifetime, is exquisitely sensitive to changes in the local environment of the fluorophore, imaging the fluorescence lifetime of NADH and NADPH offers the potential for label-free monitoring of metabolic changes inside living tumors. Here, we describe the biological, photophysical, and methodological considerations required to establish fluorescence lifetime imaging (FLIM) of NAD(P)H as a routine method for profiling the metabolism of living cancer cells and tissues.
- Published
- 2019
22. Biclustering Analysis of Co-regulation Patterns in Nuclear-Encoded Mitochondrial Genes and Metabolic Pathways
- Author
-
Robert B, Bentham, Kevin, Bryson, and Gyorgy, Szabadkai
- Subjects
Genes, Mitochondrial ,Gene Expression Regulation ,Gene Expression Profiling ,Databases, Genetic ,Cluster Analysis ,Computational Biology ,Gene Regulatory Networks ,Algorithms ,Metabolic Networks and Pathways ,Mitochondria - Abstract
Transcription of a large set of nuclear-encoded genes underlies biogenesis of mitochondria, regulated by a complex network of transcription factors and co-regulators. A remarkable heterogeneity can be detected in the expression of these genes in different cell types and tissues, and the recent availability of large gene expression compendiums allows the quantification of specific mitochondrial biogenesis patterns. We have developed a method to effectively perform this task. Massively correlated biclustering (MCbiclust) is a novel bioinformatics method that has been successfully applied to identify co-regulation patterns in large genesets, underlying essential cellular functions and determining cell types. The method has been recently evaluated and made available as a package in Bioconductor for R. One of the potential applications of the method is to compare expression of nuclear-encoded mitochondrial genes or larger sets of metabolism-related genes between different cell types or cellular metabolic states. Here we describe the essential steps to use MCbiclust as a tool to investigate co-regulation of mitochondrial genes and metabolic pathways.
- Published
- 2019
23. Determination of ATP, ADP, and AMP Levels by Reversed-Phase High-Performance Liquid Chromatography in Cultured Cells
- Author
-
Michela, Menegollo, Isabella, Tessari, Luigi, Bubacco, and Gyorgy, Szabadkai
- Subjects
Adenosine Diphosphate ,Chromatography, Reverse-Phase ,Adenosine Triphosphate ,Cell Line, Tumor ,MCF-7 Cells ,Humans ,Breast Neoplasms ,Female ,Adenosine Monophosphate ,Chromatography, High Pressure Liquid - Abstract
Cytoplasmic and mitochondrial Ca
- Published
- 2019
24. CHCHD4 regulates a proliferation-EMT switch in tumour cells, through respiratory complex I mediated metabolism
- Author
-
Gyorgy Szabadkai, Christian Frezza, Ana S. H. Costa, Cinzia Esposito, Margaret Ashcroft, Thomas S. Blacker, Jenna M. Stephen, and Luke W. Thomas
- Subjects
0303 health sciences ,biology ,Cell growth ,Chemistry ,Respiratory chain complex ,Respiratory chain ,Vimentin ,mTORC1 ,Gene signature ,Hedgehog signaling pathway ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,biology.protein ,Intracellular ,030304 developmental biology - Abstract
BACKGROUNDMitochondrial metabolism involves oxidative phosphorylation (OXPHOS) via the respiratory chain and is required for the maintenance of tumour cell proliferation and regulation of epithelial–mesenchymal transition (EMT)-related phenotypes through mechanisms that are not fully understood. The essential mitochondrial import protein coiled-coil helix coiled-coil helix domain-containing protein 4 (CHCHD4) controls respiratory chain complex activity and oxygen consumption, and regulates the growth of tumours in vivo. In this study we interrogate the role of CHCHD4-regulated respiratory chain activity and metabolism in tumour cell proliferation and EMT-related phenotypes.RESULTSWe show that CHCHD4 is essential for the proliferation of tumour cells irrespective of their aetiology. In human tumours, elevated CHCHD4 expression is correlated with a mitochondrial OXPHOS gene signature and with a proliferative gene signature associated with the mTORC1 signalling pathway. Elevated CHCHD4 increases tumour cell proliferation, in a manner that is dependent on complex I (CI) activity, glutamine consumption and mTORC1 activation. CHCHD4 expression is inversely correlated with EMT gene expression both in vitro and in vivo. Finally, we show CHCHD4 regulates the intracellular distribution of the EMT marker vimentin, in a CI-mediated manner.CONCLUSIONSCHCHD4 regulates tumour cell proliferation and metastatic (EMT-related) phenotypes through its control of CI-mediated mitochondrial metabolism.
- Published
- 2019
- Full Text
- View/download PDF
25. Metabolic Profiling of Live Cancer Tissues Using NAD(P)H Fluorescence Lifetime Imaging
- Author
-
Gyorgy Szabadkai, Michael R. Duchen, Michael D E Sewell, and Thomas S. Blacker
- Subjects
Autofluorescence ,Cancer metabolism ,Fluorescence lifetime imaging ,Live-cell microscopy ,NADH ,NADPH ,Data Analysis ,Energy Metabolism ,Humans ,Liver Neoplasms ,Microscopy, Fluorescence ,NADP ,Research Design ,Metabolome ,Metabolomics ,Optical Imaging ,0301 basic medicine ,Fluorescence-lifetime imaging microscopy ,Fluorophore ,Fluorescence ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Microscopy ,Metabolism ,Metabolic pathway ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Biophysics ,NAD+ kinase - Abstract
Altered metabolism is a hallmark of cancer, both resulting from and driving oncogenesis. The NAD and NADP redox couples play a key role in a large number of the metabolic pathways involved. In their reduced forms, NADH and NADPH, these molecules are intrinsically fluorescent. As the average time for fluorescence to be emitted following excitation by a laser pulse, the fluorescence lifetime, is exquisitely sensitive to changes in the local environment of the fluorophore, imaging the fluorescence lifetime of NADH and NADPH offers the potential for label-free monitoring of metabolic changes inside living tumors. Here, we describe the biological, photophysical, and methodological considerations required to establish fluorescence lifetime imaging (FLIM) of NAD(P)H as a routine method for profiling the metabolism of living cancer cells and tissues.
- Published
- 2019
26. Biclustering Analysis of Co-regulation Patterns in Nuclear-Encoded Mitochondrial Genes and Metabolic Pathways
- Author
-
Gyorgy Szabadkai, Kevin Bryson, and Robert B Bentham
- Subjects
Mitochondrial DNA ,Cell type ,MCbiclust ,Computational biology ,Mitochondrion ,Biology ,Databases ,03 medical and health sciences ,0302 clinical medicine ,Genetic ,Gene expression ,Cluster Analysis ,Gene Regulatory Networks ,Biclustering ,Metabolism ,Mitochondria ,Algorithms ,Databases, Genetic ,Metabolic Networks and Pathways ,Computational Biology ,Gene Expression Profiling ,Gene Expression Regulation ,Genes, Mitochondrial ,Transcription factor ,Gene ,030304 developmental biology ,0303 health sciences ,Mitochondrial ,Genes ,Mitochondrial biogenesis ,030217 neurology & neurosurgery ,Biogenesis - Abstract
Transcription of a large set of nuclear-encoded genes underlies biogenesis of mitochondria, regulated by a complex network of transcription factors and co-regulators. A remarkable heterogeneity can be detected in the expression of these genes in different cell types and tissues, and the recent availability of large gene expression compendiums allows the quantification of specific mitochondrial biogenesis patterns. We have developed a method to effectively perform this task. Massively correlated biclustering (MCbiclust) is a novel bioinformatics method that has been successfully applied to identify co-regulation patterns in large genesets, underlying essential cellular functions and determining cell types. The method has been recently evaluated and made available as a package in Bioconductor for R. One of the potential applications of the method is to compare expression of nuclear-encoded mitochondrial genes or larger sets of metabolism-related genes between different cell types or cellular metabolic states. Here we describe the essential steps to use MCbiclust as a tool to investigate co-regulation of mitochondrial genes and metabolic pathways.
- Published
- 2019
27. CHCHD4 regulates tumour proliferation and EMT-related phenotypes, through respiratory chain-mediated metabolism
- Author
-
Gyorgy Szabadkai, Margaret Ashcroft, Cinzia Esposito, Christian Frezza, Jenna M. Stephen, Luke W. Thomas, Ana S. H. Costa, Thomas S. Blacker, Ashcroft, Margaret [0000-0002-0066-3707], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cell ,respiratory chain ,Respiratory chain ,HIF-1α ,mTORC1 ,Mitochondrion ,Biology ,Coiled-coil helix coiled-coil helix domain-containing protein 4 (CHCHD4) ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,complex I ,disulfide relay system ,hypoxia ,mitochondria ,tumour growth ,tumour metabolism ,Gene expression ,medicine ,Cell growth ,Research ,Respiratory chain complex ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Cell biology ,Psychiatry and Mental health ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis - Abstract
Background Mitochondrial oxidative phosphorylation (OXPHOS) via the respiratory chain is required for the maintenance of tumour cell proliferation and regulation of epithelial to mesenchymal transition (EMT)-related phenotypes through mechanisms that are not fully understood. The essential mitochondrial import protein coiled-coil helix coiled-coil helix domain-containing protein 4 (CHCHD4) controls respiratory chain complex activity and oxygen consumption, and regulates the growth of tumours in vivo. In this study, we interrogate the importance of CHCHD4-regulated mitochondrial metabolism for tumour cell proliferation and EMT-related phenotypes, and elucidate key pathways involved. Results Using in silico analyses of 967 tumour cell lines, and tumours from different cancer patient cohorts, we show that CHCHD4 expression positively correlates with OXPHOS and proliferative pathways including the mTORC1 signalling pathway. We show that CHCHD4 expression significantly correlates with the doubling time of a range of tumour cell lines, and that CHCHD4-mediated tumour cell growth and mTORC1 signalling is coupled to respiratory chain complex I (CI) activity. Using global metabolomics analysis, we show that CHCHD4 regulates amino acid metabolism, and that CHCHD4-mediated tumour cell growth is dependent on glutamine. We show that CHCHD4-mediated tumour cell growth is linked to CI-regulated mTORC1 signalling and amino acid metabolism. Finally, we show that CHCHD4 expression in tumours is inversely correlated with EMT-related gene expression, and that increased CHCHD4 expression in tumour cells modulates EMT-related phenotypes. Conclusions CHCHD4 drives tumour cell growth and activates mTORC1 signalling through its control of respiratory chain mediated metabolism and complex I biology, and also regulates EMT-related phenotypes of tumour cells. Electronic supplementary material The online version of this article (10.1186/s40170-019-0200-4) contains supplementary material, which is available to authorized users.
- Published
- 2019
28. Mitochondria Form Contact Sites with the Nucleus to Couple Pro-Survival Retrograde Response
- Author
-
J. Paul Chapple, Gyorgy Szabadkai, Maria Soledad Alvarez, Xia Dong, Lisa E.L. Romano, Rosella Abeti, Marta Mainenti, Robert B Bentham, Roland A. Fleck, Radha Desai, Liana Hardy, Daniel A. East, Laura Kuhlman Hussey, Anne Hamechar-Brady, Ken C. Smith, James Crosby, Valentina Zappulli, Aarti Singh, Manuel Rigon, Danilo Faccenda, Michelangelo Campanella, Gema Vizcay-Barrena, and Gurtej K. Dhoot
- Subjects
Cellular adaptation ,biology ,Chemistry ,Adaptive response ,Mitochondrion ,Cell biology ,medicine.anatomical_structure ,medicine ,Translocator protein ,biology.protein ,Retrograde signaling ,Nucleus ,Transcription factor ,Function (biology) - Abstract
Mitochondria drive cellular adaptation to stress by retro-communicating with the nucleus to promote cytoprotective mechanisms. This process of inter-organelles interplay is known as the Mitochondrial Retrograde Response (MRR) and is enacted by alterations in the mitochondrial function which result into the nuclear stabilization of transcription factors such as the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Here, we demonstrate that this adaptive response is relying on the formation of contact sites between mitochondria and nucleus. These hot-spots of communication allow for the redistribution of cholesterol to the nucleus which favours nuclear residency and activity of the NF-kB. The expression level of the cholesterol Translocator Protein (TSPO), resident on the Outer Mitochondrial Membrane (OMM), is key to the formation of these contact sites and the implementation of the cytoprotective response. This work provides therefore a new paradigm in the intracellular communication during stress by reporting the first evidence for a functional and biochemical tethering between mitochondria and nucleus which governs cellular adaptation.
- Published
- 2019
29. Additional file 4: of CHCHD4 regulates tumour proliferation and EMT-related phenotypes, through respiratory chain-mediated metabolism
- Author
-
Thomas, Luke, Esposito, Cinzia, Stephen, Jenna, Costa, Ana, Frezza, Christian, Blacker, Thomas, Gyorgy Szabadkai, and Ashcroft, Margaret
- Abstract
Figure S4. CHCHD4-mediated tumour cell growth is linked to CI-regulated mTORC1 signalling and amino acid metabolism. Chart shows extracellular levels of glutamine measured in culture medium from control U2OS cells, and cells expressing wild-type CHCHD4 (WT.cl1). Representative of 2 experiments. ±SD. n = 5. (PDF 61 kb)
- Published
- 2019
- Full Text
- View/download PDF
30. Determination of ATP, ADP, and AMP Levels by Reversed-Phase High-Performance Liquid Chromatography in Cultured Cells
- Author
-
Michela Menegollo, Luigi Bubacco, Gyorgy Szabadkai, and Isabella Tessari
- Subjects
Ca ,Bioenergetics ,2+ ,Oxidative phosphorylation ,01 natural sciences ,High-performance liquid chromatography ,ATP/ADP ratio ,03 medical and health sciences ,Adenine nucleotide ,Glycolysis ,Nucleotide ,Energy charge ,030304 developmental biology ,chemistry.chemical_classification ,Reversed-phase high-performance liquid chromatography (RP-HPLC) ,0303 health sciences ,Chemistry ,010401 analytical chemistry ,signal ,0104 chemical sciences ,Biophysics ,ATP–ADP translocase - Abstract
Cytoplasmic and mitochondrial Ca2+ signals couple cellular ATP production to activity-related energy demand. In order to accurately determine the bioenergetic effect of Ca2+ signals, cellular energy charge, i.e., the compound ratio of the phosphorylated adenine nucleotides AMP, ADP, and ATP, should be estimated. Reversed-phase high-performance liquid chromatography (RP-HPLC) allows the rapid separation and quantitation of these molecules. Here we describe a protocol applied in our laboratories to quantify ATP, ADP, and AMP nucleotides in cellular extracts.
- Published
- 2019
31. Mitochondrial Permeability Transition: A Molecular Lesion with Multiple Drug Targets
- Author
-
Gyorgy Szabadkai, David L. Selwood, Thomas Briston, and Michael R. Duchen
- Subjects
0301 basic medicine ,Drug ,Programmed cell death ,media_common.quotation_subject ,Disease ,Mitochondrion ,Toxicology ,mPTP ,Mitochondrial Membrane Transport Proteins ,drug discovery ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Medicine ,Animals ,Humans ,cardiovascular diseases ,Molecular Targeted Therapy ,Child ,Molecular lesion ,media_common ,Pharmacology ,calcium ,Cell Death ,business.industry ,Drug discovery ,Mitochondrial Permeability Transition Pore ,cyclophilin D ,mitochondria ,Drug Discovery ,Mitochondria ,Neurodegenerative Diseases ,Neuromuscular Diseases ,Reperfusion Injury ,MPTP ,030104 developmental biology ,chemistry ,Mitochondrial permeability transition pore ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Mitochondrial permeability transition, as the consequence of opening of a mitochondrial permeability transition pore (mPTP), is a cellular catastrophe. Initiating bioenergetic collapse and cell death, it has been implicated in the pathophysiology of major human diseases, including neuromuscular diseases of childhood, ischaemia-reperfusion injury, and age-related neurodegenerative disease. Opening of the mPTP represents a major therapeutic target, as it can be mitigated by a number of compounds. However, clinical studies have so far been disappointing. We therefore address the prospects and challenges faced in translating in vitro findings to clinical benefit. We review the role of mPTP opening in disease, discuss recent findings defining the putative structure of the mPTP, and explore strategies to identify novel, clinically useful mPTP inhibitors, highlighting key considerations in the drug discovery process.
- Published
- 2019
32. The mitochondrial calcium uniporter regulates breast cancer progression via HIF‐1α
- Author
-
Antonio Rosato, Robert B Bentham, Ivan Bogeski, Rosario Rizzuto, Michael R. Duchen, Tunde Berecz, Thomas S. Blacker, Anna Tosatto, Roberta Sommaggio, Cristina Mammucari, Carsten Kummerow, and Gyorgy Szabadkai
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Motility ,HIF-1α ,Triple Negative Breast Neoplasms ,Mice, SCID ,Biology ,Metastasis ,mitochondrial Ca2+ uptake ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Downregulation and upregulation ,Cell Movement ,Internal medicine ,Cell Line, Tumor ,medicine ,Gene silencing ,metastasis ,Animals ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Neoplasm Metastasis ,Mitochondrial Ca2+ uptake ,Reactive oxygen species ,Molecular Medicine ,Research Articles ,Cancer ,Cell Proliferation ,reactive oxygen species ,Cell growth ,HIF‐1α ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular medicine ,030104 developmental biology ,Endocrinology ,Metabolism ,030220 oncology & carcinogenesis ,Cancer research ,Heterografts ,Female ,Calcium Channels ,Research Article - Abstract
Triple‐negative breast cancer (TNBC) represents the most aggressive breast tumor subtype. However, the molecular determinants responsible for the metastatic TNBC phenotype are only partially understood. We here show that expression of the mitochondrial calcium uniporter (MCU), the selective channel responsible for mitochondrial Ca2+ uptake, correlates with tumor size and lymph node infiltration, suggesting that mitochondrial Ca2+ uptake might be instrumental for tumor growth and metastatic formation. Accordingly, MCU downregulation hampered cell motility and invasiveness and reduced tumor growth, lymph node infiltration, and lung metastasis in TNBC xenografts. In MCU‐silenced cells, production of mitochondrial reactive oxygen species (mROS) is blunted and expression of the hypoxia‐inducible factor‐1α (HIF‐1α) is reduced, suggesting a signaling role for mROS and HIF‐1α, downstream of mitochondrial Ca2+. Finally, in breast cancer mRNA samples, a positive correlation of MCU expression with HIF‐1α signaling route is present. Our results indicate that MCU plays a central role in TNBC growth and metastasis formation and suggest that mitochondrial Ca2+ uptake is a potential novel therapeutic target for clinical intervention.
- Published
- 2016
33. Correction: Impaired cellular bioenergetics caused by GBA1 depletion sensitizes neurons to calcium overload
- Author
-
Dany P. Perocheau, Nicoletta Plotegher, Gauri Bhosale, Michael R. Duchen, Giulia Massaro, Graziano Guella, Federico Zambon, Ruggero Ferrazza, Gyorgy Szabadkai, Simon N. Waddington, and Ahad A. Rahim
- Subjects
Cellular bioenergetics ,Chemistry ,Cell Biology ,Calcium overload ,Molecular Biology ,Cell biology - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
34. Mitochondria form contact sites with the nucleus to couple pro-survival retrograde response
- Author
-
Michelangelo Campanella, Roland A. Fleck, Radha Desai, Liana Hardy, Gyorgy Szabadkai, J. Paul Chapple, Ken C. Smith, Manuel Rigon, Anne Hamechar-Brady, Valentina Zappulli, Robert B Bentham, Gema Vizcay-Barrena, Danilo Faccenda, Lisa E.L. Romano, Gurtej K. Dhoot, Marta Mainenti, Maria Soledad Alvarez, James Crosby, Arti Singh, Laura Kuhlman Hussey, Daniel A. East, and Xia Dong
- Subjects
biology ,Cholesterol ,Cellular homeostasis ,Mitochondrion ,Cell biology ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,medicine ,Retrograde signaling ,Translocator protein ,biology.protein ,Nucleus ,Reprogramming ,Hormone - Abstract
Mitochondria drive cellular adaptation to stress by retro-communicating with the nucleus. This process is known as Mitochondrial Retrograde Response (MRR) and is induced by mitochondrial dysfunctions which perturb cell signalling. MRR results in the nuclear stabilization and activation of pro-survival transcription factors such as the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Here we demonstrate that MRR is facilitated by the formation of contact sites between mitochondria and the nucleus which establish microdomains of communication between the two organelles. The 18kD Translocator Protein (TSPO), which de-ubiquitylates and stabilizes the mitochondrial network preventing its mitophagy-mediated segregation, is required for this interaction. The tethering TSPO enacts is mediated by the complex formed with the Protein Kinase A via the A-kinase anchoring protein Acyl-CoA Binding Domain Containing 3 (ACBD3) and allows the redistribution of cholesterol which sustains the pro-survival response by blocking NF-kB de-acetylation. This work proposes a new paradigm in the mitochondrial retro-communication by revealing the existence of contact sites between mitochondrial and the nucleus and a signalling role for cholesterol.
- Published
- 2018
35. Fantastic beasts and how to find them—Molecular identification of the mitochondrial ATP-sensitive potassium channel
- Author
-
Gyorgy Szabadkai, Sean M. Davidson, and Michael R. Duchen
- Subjects
0301 basic medicine ,ATP-sensitive potassium channel ,Physiology ,Potassium ,chemistry.chemical_element ,Mitochondrion ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,KATP Channels ,Katp channels ,Potassium Channel Blockers ,Animals ,Humans ,Molecular Biology ,Molecular identification ,Ischaemia and reperfusion ,Chemistry ,Mitochondrial calcium uniporter ,Cell Biology ,Mitochondria ,Katp channel ,030104 developmental biology ,Reperfusion Injury ,Biophysics ,030217 neurology & neurosurgery ,Communication channel - Abstract
Despite reported sightings over many years, certain mitochondrial-specific channels have proven to be elusive beasts, evading molecular identification. However, combining modern genetics with a wave of their ion-sensing wand, researchers have managed to capture first the mitochondrial calcium uniporter, and now that semi-mythological beast, the mitochondrial ATP-sensitive potassium (mitoKATP) channel.
- Published
- 2019
36. Targeting RORs nuclear receptors by novel synthetic steroidal inverse agonists for autoimmune disorders
- Author
-
Davide Carta, Yahima Frión-Herrera, Matteo Suman, Nicola Paccagnella, Matteo Dal Pra, Giulia Castellani, Maria Grazia Ferlin, Gyorgy Szabadkai, and Sara De Martin
- Subjects
0301 basic medicine ,Drug Inverse Agonism ,Cell Survival ,Receptors, Retinoic Acid ,Clinical Biochemistry ,Pharmaceutical Science ,Autoimmune disorders ,Gal4 UAS-Luc co-transfection ,Inverse agonists ,NR RORγt ,Biochemistry ,Molecular Medicine ,Molecular Biology ,3003 ,Drug Discovery3003 Pharmaceutical Science ,Organic Chemistry ,Autoimmune Diseases ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Ursolic acid ,RAR-related orphan receptor gamma ,Drug Discovery ,Tumor Cells, Cultured ,Humans ,Inverse agonist ,Potency ,Viability assay ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Cell Cycle ,Hep G2 Cells ,Molecular Docking Simulation ,HEK293 Cells ,030104 developmental biology ,Nuclear receptor ,Steroids ,Lead compound ,Derivative (chemistry) - Abstract
Designing novel inverse agonists of NR RORγt still represents a challenge for the pharmaceutical community to develop therapeutics for treating immune diseases. By exploring the structure of NRs natural ligands, the representative arotenoid ligands and RORs specific ligands share some chemical homologies which can be exploited to design a novel molecular structure characterized by a polycyclic core bearing a polar head and a hydrophobic tail. Compound MG 2778 (8), a cyclopenta[a]phenantrene derivative, was identified as lead compound which was chemically modified at position 2 in order to obtain a small library for preliminary SARs. Cell viability and estrogenic activity of compounds 7, 8, 19a, 30, 31 and 32 were evaluated to attest selectivity. The selected 7, 8, 19a and 31 compounds were assayed in a Gal4 UAS-Luc co-transfection system in order to determine their ability to modulate RORγt activity in a cellular environment. They were evaluated as inverse agonists taken ursolic acid as reference compound. The potency of compounds was lower than that of ursolic acid, but their efficacy was similar. Compound 19a was the most active, significantly reducing RORγt activity at low micromolar concentrations.
- Published
- 2018
37. Inositol trisphosphate receptor-mediated Ca 2+ signalling stimulates mitochondrial function and gene expression in core myopathy patients
- Author
-
Matteo, Suman, Jenny A, Sharpe, Robert B, Bentham, Vassilios N, Kotiadis, Michela, Menegollo, Viviana, Pignataro, Jordi, Molgó, Francesco, Muntoni, Michael R, Duchen, Elena, Pegoraro, and Gyorgy, Szabadkai
- Subjects
Muscle Fibers, Skeletal ,Animals ,Calcium Signaling ,Gene Expression Regulation ,Humans ,Inositol ,Inositol 1,4,5-Trisphosphate Receptors ,Mice ,Mitochondria ,Muscle, Skeletal ,Muscular Diseases ,Mutation ,Ryanodine Receptor Calcium Release Channel ,Molecular Biology ,Genetics ,Genetics (clinical) ,Skeletal ,Inositol 1 ,Muscle Fibers ,Muscle ,5-Trisphosphate Receptors - Abstract
Core myopathies are a group of childhood muscle disorders caused by mutations of the ryanodine receptor (RyR1), the Ca2+ release channel of the sarcoplasmic reticulum. These mutations have previously been associated with elevated inositol trisphosphate receptor (IP3R) levels in skeletal muscle myotubes derived from patients. However, the functional relevance and the relationship of IP3R mediated Ca2+ signalling with the pathophysiology of the disease is unclear. It has also been suggested that mitochondrial dysfunction underlies the development of central and diffuse multi-mini-cores, devoid of mitochondrial activity, which is a key pathological consequence of RyR1 mutations. Here we used muscle biopsies of central core and multi-minicore disease patients with RyR1 mutations, as well as cellular and in vivo mouse models of the disease to characterize global cellular and mitochondrial Ca2+ signalling, mitochondrial function and gene expression associated with the disease. We show that RyR1 mutations that lead to the depletion of the channel are associated with increased IP3-mediated nuclear and mitochondrial Ca2+ signals and increased mitochondrial activity. Moreover, western blot and microarray analysis indicated enhanced mitochondrial biogenesis at the transcriptional and protein levels and was reflected in increased mitochondrial DNA content. The phenotype was recapitulated by RYR1 silencing in mouse cellular myotube models. Altogether, these data indicate that remodelling of skeletal muscle Ca2+ signalling following loss of functional RyR1 mediates bioenergetic adaptation.
- Published
- 2018
38. Mitochondria and Reactive Oxygen Species in Aging and Age-Related Diseases
- Author
-
Pawel Dobrzyn, Piero Portincasa, Barbara Zavan, Massimo Bonora, Paulo J. Oliveira, Giampaolo Morciano, Sabine Borchard, Hans Zischka, Gyorgy Szabadkai, Alessandro Rimessi, Jerzy Duszyński, Grzegorz Węgrzyn, Agnieszka Dobrzyn, Paulina Patalas-Krawczyk, Jędrzej Szymański, Karolina Pierzynowska, David Q.-H. Wang, Vilma A. Sardão, Carlotta Giorgi, Saverio Marchi, Paulina Jędrak, Agnieszka Karkucinska-Wieckowska, Ines C.M. Simoes, Ziyu Ren, Mariusz R. Wieckowski, Paolo Pinton, and Mariasole Perrone
- Subjects
0301 basic medicine ,Mitochondrial ROS ,Aging ,Cell ,Socio-culturale ,Mitochondrion ,Biology ,Biochemistry ,Article ,Pathogenesis ,03 medical and health sciences ,Anti-ROS intervention ,Antioxidant defense ,Gene expression ,Organelle ,medicine ,Animals ,Humans ,Mitochondrial dysfunction–related pathologies ,Molecular Biology ,chemistry.chemical_classification ,Reactive oxygen species ,Age-related Neurodegenerative Disorders ,Anti-ros Intervention ,Antioxidant Defense ,Mitochondria ,Mitochondrial Dysfunction–related Pathologies ,Ros ,Age-related neurodegenerative disorders ,Eukaryota ,ROS ,Cell Biology ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Signal transduction ,Energy Metabolism ,Reactive Oxygen Species ,Signal Transduction - Abstract
Aging has been linked to several degenerative processes that, through the accumulation of molecular and cellular damage, can progressively lead to cell dysfunction and organ failure. Human aging is linked with a higher risk for individuals to develop cancer, neurodegenerative, cardiovascular, and metabolic disorders. The understanding of the molecular basis of aging and associated diseases has been one major challenge of scientific research over the last decades. Mitochondria, the center of oxidative metabolism and principal site of reactive oxygen species (ROS) production, are crucial both in health and in pathogenesis of many diseases. Redox signaling is important for the modulation of cell functions and several studies indicate a dual role for ROS in cell physiology. In fact, high concentrations of ROS are pathogenic and can cause severe damage to cell and organelle membranes, DNA, and proteins. On the other hand, moderate amounts of ROS are essential for the maintenance of several biological processes, including gene expression. In this review, we provide an update regarding the key roles of ROS—mitochondria cross talk in different fundamental physiological or pathological situations accompanying aging and highlighting that mitochondrial ROS may be a decisive target in clinical practice.
- Published
- 2018
- Full Text
- View/download PDF
39. NADH Shuttling Couples Cytosolic Reductive Carboxylation of Glutamine with Glycolysis in Cells with Mitochondrial Dysfunction
- Author
-
Edoardo Gaude, Christina Schmidt, Payam A. Gammage, Aurelien Dugourd, Thomas Blacker, Sew Peak Chew, Julio Saez-Rodriguez, John S. O’Neill, Gyorgy Szabadkai, Michal Minczuk, Christian Frezza
- Published
- 2018
- Full Text
- View/download PDF
40. The regulation of neuronal mitochondrial metabolism by calcium
- Author
-
Gyorgy Szabadkai, Michael R. Duchen, Jorgina Satrústegui, Carlos B. Rueda, Beatriz Pardo, and Irene Llorente-Folch
- Subjects
medicine.medical_specialty ,biology ,Physiology ,Malate-aspartate shuttle ,chemistry.chemical_element ,Mitochondrion ,Calcium ,Cell biology ,Citric acid cycle ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,medicine ,Glutamate aspartate transporter ,biology.protein ,Veratridine ,Inner mitochondrial membrane ,Calcium signaling - Abstract
Calcium signalling is fundamental to the function of the nervous system, in association with changes in ionic gradients across the membrane. Although restoring ionic gradients is energetically costly, a rise in intracellular Ca(2+) acts through multiple pathways to increase ATP synthesis, matching energy supply to demand. Increasing cytosolic Ca(2+) stimulates metabolite transfer across the inner mitochondrial membrane through activation of Ca(2+) -regulated mitochondrial carriers, whereas an increase in matrix Ca(2+) stimulates the citric acid cycle and ATP synthase. The aspartate-glutamate exchanger Aralar/AGC1 (Slc25a12), a component of the malate-aspartate shuttle (MAS), is stimulated by modest increases in cytosolic Ca(2+) and upregulates respiration in cortical neurons by enhancing pyruvate supply into mitochondria. Failure to increase respiration in response to small (carbachol) and moderate (K(+) -depolarization) workloads and blunted stimulation of respiration in response to high workloads (veratridine) in Aralar/AGC1 knockout neurons reflect impaired MAS activity and limited mitochondrial pyruvate supply. In response to large workloads (veratridine), acute stimulation of respiration occurs in the absence of MAS through Ca(2+) influx through the mitochondrial calcium uniporter (MCU) and a rise in matrix [Ca(2+) ]. Although the physiological importance of the MCU complex in work-induced stimulation of respiration of CNS neurons is not yet clarified, abnormal mitochondrial Ca(2+) signalling causes pathology. Indeed, loss of function mutations in MICU1, a regulator of MCU complex, are associated with neuromuscular disease. In patient-derived MICU1 deficient fibroblasts, resting matrix Ca(2+) is increased and mitochondria fragmented. Thus, the fine tuning of Ca(2+) signals plays a key role in shaping mitochondrial bioenergetics.
- Published
- 2015
41. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015
- Author
-
Lorenzo Galluzzi, Thomas Rudel, Hans-Uwe Simon, Vishva M. Dixit, Erwin F. Wagner, Marie-Lise Gougeon, Andreas Linkermann, J M Bravo-San Pedro, Rosario Rizzuto, Cecília M. P. Rodrigues, Gian Maria Fimia, Hidenori Ichijo, Mathieu J.M. Bertrand, Kodi S. Ravichandran, Francis Ka-Ming Chan, Stephen W.G. Tait, Jochen H. M. Prehn, Richard A. Lockshin, Valina L. Dawson, Andreas Villunger, Sharad Kumar, Emily H. Cheng, Carlos López-Otín, Theocharis Panaretakis, Lucia Altucci, Gabriel A. Rabinovich, Michelangelo Campanella, Peter Vandenabeele, Marcus E. Peter, Francesco Cecconi, Noboru Mizushima, Ilio Vitale, Frank Madeo, Mikhail V. Blagosklonny, Zahra Zakeri, Stuart A. Aaronson, Gabriel Núñez, Eric H. Baehrecke, Nektarios Tavernarakis, Gyorgy Szabadkai, Eleonora Candi, Brent R. Stockwell, Dale E. Bredesen, Seamus J. Martin, Thomas Kaufmann, Sonia Melino, Dieter Adam, John M. Abrams, Katiuscia Bianchi, Yufang Shi, Emad S. Alnemri, Klas Blomgren, Pascal Meier, Catherine Brenner, Michael O. Hengartner, Philipp J. Jost, J M Hardwick, Eileen White, T Vanden Berghe, N. Di Daniele, Nicolas G. Bazan, H. L. Tang, Mauro Piacentini, V De Laurenzi, Beth Levine, Margherita Annicchiarico-Petruzzelli, Josef M. Penninger, Walter Malorni, Ted M. Dawson, Carmen Garrido, David W. Andrews, Douglas R. Green, György Hajnóczky, Jerry E. Chipuk, Wafik S. El-Deiry, Christoph Borner, Stuart A. Lipton, John A. Cidlowski, Klaus-Michael Debatin, Junying Yuan, Jan Paul Medema, Bertrand Joseph, Aaron Ciechanover, Ute M. Moll, Hinrich Gronemeyer, Paolo Pinton, Gerry Melino, Daniel J. Klionsky, Simone Fulda, John J. Lemasters, Cristina Muñoz-Pinedo, Hamsa Puthalakath, Navdeep S. Chandel, R De Maria, Jean-Christophe Marine, Richard A. Flavell, Brian David Dynlacht, W. G. Wood, Henning Walczak, David C. Rubinsztein, Guido Kroemer, Oliver Kepp, Richard A. Knight, Andrew Oberst, Enrico Lugli, J-C Martinou, Boris Zhivotovsky, Yoshihide Tsujimoto, Galluzi, L, Bravo-San, Pedro JM, Vitale, I, Aaaronson, SA, Kumar, S, Kroemer, Guido, Galluzzi, L, Bravo San Pedro, J. M, Aaronson, S. A, Abrams, J. M, Adam, D, Alnemri, E. S, Altucci, L, Andrews, D, Annicchiarico Petruzzelli, M, Baehrecke, E. H, Bazan, N. G, Bertrand, M. J, Bianchi, K, Blagosklonny, M. V, Blomgren, K, Borner, C, Bredesen, D. E, Brenner, C, Campanella, M, Candi, E, Cecconi, F, Chan, F. K, Chandel, N. S, Cheng, E. H, Chipuk, J. E, Cidlowski, J. A, Ciechanover, A, Dawson, T. M, Dawson, V. L, De Laurenzi, V, De Maria, R, Debatin, K. M, Di Daniele, N, Dixit, V. M, Dynlacht, B. D, El Deiry, W. S, Fimia, Gian Maria, Flavell, R. A, Fulda, S, Garrido, C, Gougeon, M. L, Green, D. R, Gronemeyer, H, Hajnoczky, G, Hardwick, J. M, Hengartner, M. O, Ichijo, H, Joseph, B, Jost, P. J, Kaufmann, T, Kepp, O, Klionsky, D. J, Knight, R. A, Lemasters, J. J, Levine, B, Linkermann, A, Lipton, S. A, Lockshin, R. A, López Otín, C, Lugli, E, Madeo, F, Malorni, W, Marine, J. C, Martin, S. J, Martinou, J. C, Medema, J. P, Meier, P, Melino, S, Mizushima, N, Moll, U, Muñoz Pinedo, C, Nuñez, G, Oberst, A, Panaretakis, T, Penninger, J. M, Peter, M. E, Piacentini, M, Pinton, P, Prehn, J. H, Puthalakath, H, Rabinovich, G. A, Ravichandran, K. S, Rizzuto, R, Rodrigues, C. M, Rubinsztein, D. C, Rudel, T, Shi, Y, Simon, H. U, Stockwell, B. R, Szabadkai, G, Tait, S. W, Tang, H. L, Tavernarakis, N, Tsujimoto, Y, Vanden Berghe, T, Vandenabeele, P, Villunger, A, Wagner, E. F, Walczak, H, White, E, Wood, W. G, Yuan, J, Zakeri, Z, Zhivotovsky, B, Melino, G, Kroemer, G., Bravo San Pedro, Jm, Aaronson, Sa, Abrams, Jm, Alnemri, E, Altucci, Lucia, Baehrecke, Eh, Bazan, Ng, Bertrand, Mj, Blagosklonny, Mv, Bredesen, De, Chan, Fk, Chandel, N, Cheng, Eh, Chipuk, Je, Cidlowski, Ja, Dawson, Tm, Dawson, Vl, Debatin, Km, Dixit, Vm, Dynlacht, Bd, El Deiry, W, Fimia, Gm, Flavell, Ra, Gougeon, Ml, Green, Dr, Hardwick, Jm, Hengartner, Mo, Jost, Pj, Klionsky, Dj, Knight, Ra, Lemasters, Jj, Lipton, Sa, Lockshin, Ra, Marine, Jc, Martin, Sj, Martinou, Jc, Medema, Jp, Penninger, Jm, Peter, Me, Prehn, Jh, Rabinovich, Ga, Ravichandran, K, Rodrigues, Cm, Rubinsztein, Dc, Simon, Hu, Stockwell, Br, Tait, Sw, Tang, Hl, Wagner, Ef, and Wood, Wg
- Subjects
Biochemical Manifestations of Cell Death ,ISCHEMIA-REPERFUSION INJURY ,Apoptosis ,Review ,Transduction (genetics) ,0302 clinical medicine ,CASPASE INHIBITION SWITCHES ,Animals ,Humans ,Terminology as Topic ,Signal Transduction ,610 Medicine & health ,Caspase ,TUMOR-NECROSIS-FACTOR ,0303 health sciences ,Settore BIO/17 ,biology ,Settore BIO/11 ,Neurodegeneration ,Settore BIO/13 ,APOPTOSIS ,3. Good health ,Medicina Básica ,cell death ,030220 oncology & carcinogenesis ,Morphologic Aspects of Cell Death ,Signal transduction ,DOMAIN-LIKE PROTEIN ,Intracellular ,Human ,Necroptosi ,CYTOCHROME-C RELEASE ,OUTER-MEMBRANE PERMEABILIZATION ,Programmed cell death ,CIENCIAS MÉDICAS Y DE LA SALUD ,Settore BIO/06 ,Inmunología ,CELL DEATH ,NO ,Q-VD-OPH ,03 medical and health sciences ,Settore MED/04 - PATOLOGIA GENERALE ,ddc:570 ,APOPTOSIS-INDUCING FACTOR ,MIXED LINEAGE KINASE ,medicine ,Molecular Biology ,Cell Biology ,Settore BIO/10 ,030304 developmental biology ,Animal ,Cell growth ,Apoptosi ,Biology and Life Sciences ,medicine.disease ,MITOCHONDRIAL PERMEABILITY TRANSITION ,Immunology ,biology.protein ,Neuroscience - Abstract
Cells exposed to extreme physicochemical or mechanical stimuli die in an uncontrollable manner, as a result of their immediate structural breakdown. Such an unavoidable variant of cellular demise is generally referred to as ?accidental cell death' (ACD). In most settings, however, cell death is initiated by a genetically encoded apparatus, correlating with the fact that its course can be altered by pharmacologic or genetic interventions. "Regulated cell death" (RCD) can occur as part of physiologic programs or can be activated once adaptive responses to perturbations of the extracellular or intracellular microenvironment fail. The biochemical phenomena that accompany RCD may be harnessed to classify it into a few subtypes, which often (but not always) exhibit stereotyped morphologic features. Nonetheless, efficiently inhibiting the processes that are commonly thought to cause RCD, such as the activation of executioner caspases in the course of apoptosis, does not exert true cytoprotective effects in the mammalian system, but simply alters the kinetics of cellular demise as it shifts its morphologic and biochemical correlates. Conversely, bona fide cytoprotection can be achieved by inhibiting the transduction of lethal signals in the early phases of the process, when adaptive responses are still operational. Thus, the mechanisms that truly execute RCD may be less understood, less inhibitable and perhaps more homogeneous than previously thought. Here, the Nomenclature Committee on Cell Death formulates a set of recommendations to help scientists and researchers to discriminate between essential and accessory aspects of cell death Fil: Rabinovich, Gabriel Adrian. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental (i); Argentina Fil: Nomenclature Committee on Cell Death. Equipe 11 Apoptose, Cancer et Immunité. Centre de Recherche des Cordeliers; Francia
- Published
- 2015
42. mTORC1-independent autophagy regulates receptor tyrosine kinase phosphorylation in colorectal cancer cells via an mTORC2-mediated mechanism
- Author
-
Daniel Hochhauser, James O'Prey, Aikaterini Lampada, Kevin M. Ryan, Gyorgy Szabadkai, and Paolo Salomoni
- Subjects
0301 basic medicine ,Programmed cell death ,Original Paper ,biology ,ATG5 ,Autophagy ,Cell Biology ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,BAG3 ,Receptor tyrosine kinase ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Cancer research ,biology.protein ,Humans ,Phosphorylation ,Colorectal Neoplasms ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
The intracellular autophagic degradative pathway can have a tumour suppressive or tumour-promoting role depending on the stage of tumour development. Upon starvation or targeting of oncogenic receptor tyrosine kinases (RTKs), autophagy is activated owing to the inhibition of PI3K/AKT/mTORC1 signalling pathway and promotes survival, suggesting that autophagy is a relevant therapeutic target in these settings. However, the role of autophagy in cancer cells where the PI3K/AKT/mTORC1 pathway is constitutively active remains partially understood. Here we report a role for mTORC1-independent basal autophagy in regulation of RTK activation and cell migration in colorectal cancer (CRC) cells. PI3K and RAS-mutant CRC cells display basal autophagy levels despite constitutive mTORC1 signalling, but fail to increase autophagic flux upon RTK inhibition. Inhibition of basal autophagy via knockdown of ATG7 or ATG5 leads to decreased phosphorylation of several RTKs, in particular c-MET. Internalised c-MET colocalised with LAMP1-negative, LC3-positive vesicles. Finally, autophagy regulates c-MET phosphorylation via an mTORC2-dependent mechanism. Overall, our findings reveal a previously unappreciated role of autophagy and mTORC2 in regulation of oncogenic RTK activation, with implications for understanding of cancer cell signalling.
- Published
- 2017
43. Assessment of Cellular Redox State Using NAD(P)H Fluorescence Intensity and Lifetime
- Author
-
Tunde Berecz, Gyorgy Szabadkai, Michael R. Duchen, and Thomas S. Blacker
- Subjects
0301 basic medicine ,FLIM ,Fluorescence-lifetime imaging microscopy ,Redox state ,Anabolism ,Autofluorescence ,Fluorescence lifetime ,Microscopy ,NAD(P)H ,NADH ,NADPH ,Strategy and Management ,Redox ,Industrial and Manufacturing Engineering ,Cofactor ,Article ,03 medical and health sciences ,0302 clinical medicine ,biology ,Catabolism ,Chemistry ,Mechanical Engineering ,Metals and Alloys ,Metabolism ,030104 developmental biology ,Biochemistry ,biology.protein ,NAD+ kinase ,030217 neurology & neurosurgery - Abstract
NADH and NADPH are redox cofactors, primarily involved in catabolic and anabolic metabolic processes respectively. In addition, NADPH plays an important role in cellular antioxidant defence. In live cells and tissues, the intensity of their spectrally-identical autofluorescence, termed NAD(P)H, can be used to probe the mitochondrial redox state, while their distinct enzyme-binding characteristics can be used to separate their relative contributions to the total NAD(P)H intensity using fluorescence lifetime imaging microscopy (FLIM). These protocols allow differences in metabolism to be detected between cell types and altered physiological and pathological states.
- Published
- 2017
44. DJ-1 is a redox sensitive adapter protein for high molecular weight complexes involved in regulation of catecholamine homeostasis
- Author
-
Dominik Piston, Lydia Alvarez-Erviti, Vikas Bansal, Daniela Gargano, Zhi Yao, Gyorgy Szabadkai, Mark Odell, M Rhyan Puno, Benny Björkblom, Jodi Maple-Grødem, Peter Breuer, Oliver Kaut, Jan Petter Larsen, Stefan Bonn, Simon Geir Møller, Ullrich Wüllner, Anthony H V Schapira, and Matthew E Gegg
- Subjects
Dopamine ,Cell- och molekylärbiologi ,Protein Deglycase DJ-1 ,metabolism [Parkinson Disease] ,genetics [Protein Deglycase DJ-1] ,Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) ,physiology [Oxidative Stress] ,metabolism [Adaptor Proteins, Signal Transducing] ,03 medical and health sciences ,Catecholamines ,0302 clinical medicine ,genetics [Parkinson Disease] ,ddc:570 ,Cell Line, Tumor ,Genetics ,Homeostasis ,Humans ,metabolism [Dopamine] ,Molecular Biology ,Medicinsk bioteknologi (med inriktning mot cellbiologi (inklusive stamcellsbiologi), molekylärbiologi, mikrobiologi, biokemi eller biofarmaci) ,Genetics (clinical) ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,0303 health sciences ,Intracellular Signaling Peptides and Proteins ,Brain ,Parkinson Disease ,Articles ,General Medicine ,metabolism [Catecholamines] ,metabolism [Protein Deglycase DJ-1] ,Oxidative Stress ,metabolism [Brain] ,PARK7 protein, human ,030220 oncology & carcinogenesis ,Corrigendum ,B140 Neuroscience ,metabolism [Intracellular Signaling Peptides and Proteins] ,Oxidation-Reduction ,Cell and Molecular Biology - Abstract
DJ-1 is an oxidation sensitive protein encoded by the PARK7 gene. Mutations in PARK7 are a rare cause of familial recessive Parkinson's disease (PD), but growing evidence suggests involvement of DJ-1 in idiopathic PD. The key clinical features of PD, rigidity and bradykinesia, result from neurotransmitter imbalance, particularly the catecholamines dopamine (DA) and noradrenaline. We report in human brain and human SH-SY5Y neuroblastoma cell lines that DJ-1 predominantly forms high molecular weight (HMW) complexes that included RNA metabolism proteins hnRNPA1 and PABP1 and the glycolysis enzyme GAPDH. In cell culture models the oxidation status of DJ-1 determined the specific complex composition. RNA sequencing indicated that oxidative changes to DJ-1 were concomitant with changes in mRNA transcripts mainly involved in catecholamine metabolism. Importantly, loss of DJ-1 function upon knock down (KD) or expression of the PD associated form L166P resulted in the absence of HMW DJ-1 complexes. In the KD model, the absence of DJ-1 complexes was accompanied by impairment in catecholamine homeostasis, with significant increases in intracellular DA and noraderenaline levels. These changes in catecholamines could be rescued by re-expression of DJ-1. This catecholamine imbalance may contribute to the particular vulnerability of dopaminergic and noradrenergic neurons to neurodegeneration in PARK7-related PD. Notably, oxidised DJ-1 was significantly decreased in idiopathic PD brain, suggesting altered complex function may also play a role in the more common sporadic form of the disease. Correction: Dominik Piston, Lydia Alvarez-Erviti, Vikas Bansal, Daniela Gargano, Zhi Yao, Gyorgy Szabadkai, Mark Odell, M Rhyan Puno, Benny Björkblom, Jodi Maple-Grødem, Peter Breuer, Oliver Kaut, Jan Petter Larsen, Stefan Bonn, Simon Geir Møller, Ullrich Wüllner, Anthony H V Schapira, Matthew E Gegg. DJ-1 is a redox sensitive adapter protein for high molecular weight complexes involved in regulation of catecholamine homeostasis. Human Molecular Genetics. Volume 27, Issue 3, 1 February 2018, Pages 576. DOI: 10.1093/hmg/ddx425
- Published
- 2017
45. Measurement of ATP in Single Oocytes: Impact of Maturation and Cumulus Cells on Levels and Consumption
- Author
-
John L. Carroll, Caroline Dalton, and Gyorgy Szabadkai
- Subjects
Cell signaling ,Time Factors ,Physiology ,In Vitro Oocyte Maturation Techniques ,Clinical Biochemistry ,Biosensing Techniques ,Cell Communication ,Fertilization in Vitro ,Biology ,Mitochondrion ,Time-Lapse Imaging ,law.invention ,Mice ,Adenosine Triphosphate ,law ,Fluorescence Resonance Energy Transfer ,medicine ,Animals ,Cells, Cultured ,Cumulus Cells ,Gap Junctions ,Embryo ,Cell Biology ,Oocyte ,Coculture Techniques ,Cell biology ,Cytosol ,medicine.anatomical_structure ,Oocytes ,Recombinant DNA ,Female ,Extended time ,Energy Metabolism - Abstract
Mitochondria provide the primary source of ATP in the oocyte and early embryo and mitochondrial dysfunction and deficit of mitochondria-derived ATP has been linked to suboptimal developmental competence. We have undertaken a study of ATP in the maturing mouse oocyte using a novel recombinant FRET based probe, AT1.03. We show that AT1.03 can be successfully used to monitor cytosolic ATP levels in single live oocytes over extended time periods. We find that ATP levels undergo dynamic changes associated with specific maturational events and that oocytes display altered rates of ATP consumption at different stages of maturation. Cumulus enclosed oocytes have a higher ATP level during maturation than denuded oocytes and this can be abolished by inhibition of gap junctional communication between the oocyte and cumulus cells. Our work uses a new approach to shed light on regulation of ATP levels and ATP consumption during oocyte maturation.
- Published
- 2013
46. Kαλóς και Aγαθóς: how mitochondrial beauty translates into biological virtue
- Author
-
Gyorgy Szabadkai and Rosario Rizzuto
- Subjects
Virtue ,media_common.quotation_subject ,Physiology ,Cell Biology ,Biology ,Endoplasmic Reticulum ,Cell Physiological Phenomena ,Mitochondria ,Evolutionary biology ,Beauty ,Animals ,Humans ,Calcium Signaling ,media_common - Abstract
We are currently witnessing a stream of major discoveries describing the elementary constituents of complex mitochondrial machineries that define the shape, dynamics and intracellular distribution of the organelle. On the basis of these findings the next wave of research holds the promise of unveiling the so far hidden relationships between the fascinating ultrastructural mitochondrial anatomy and its key roles in many areas of cell biology.
- Published
- 2013
47. Altered Ca2+ Homeostasis and Endoplasmic Reticulum Stress in Myotonic Dystrophy Type 1 Muscle Cells
- Author
-
Emanuele Loro, Matteo Suman, Annalisa Botta, Boris Pantic, Lodovica Vergani, Adriana Malena, Gyorgy Szabadkai, and Giulia Del Moro
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,SercA1 ,SERCA ,lcsh:QH426-470 ,Ca2+ homeostasis ,Biology ,Myotonic dystrophy ,Article ,Internal medicine ,Ryr1 ,Genetics ,medicine ,Myotonic dystrophy, muscle cells, Ca2+ homeostasis, SercA1, Ryr1 ,Myocyte ,Genetics (clinical) ,myotonic dystrophy ,Ryanodine receptor ,Myogenesis ,Endoplasmic reticulum ,Skeletal muscle ,muscle cells ,medicine.disease ,Cell biology ,lcsh:Genetics ,Endocrinology ,medicine.anatomical_structure ,Settore MED/03 - Genetica Medica ,Unfolded protein response ,Cav1.1 ,ER stress - Abstract
The pathogenesis of Myotonic Dystrophy type 1 (DM1) is linked to unstable CTG repeats in the DMPK gene which induce the mis-splicing to fetal/neonatal isoforms of many transcripts, including those involved in cellular Ca2+ homeostasis. Here we monitored the splicing of three genes encoding for Ca2+ transporters and channels (RyR1, SERCA1 and CACN1S) during maturation of primary DM1 muscle cells in parallel with the functionality of the Excitation-Contraction (EC) coupling machinery. At 15 days of differentiation, fetal isoforms of SERCA1 and CACN1S mRNA were significantly higher in DM1 myotubes compared to controls. Parallel functional studies showed that the cytosolic Ca2+ response to depolarization in DM1 myotubes did not increase during the progression of differentiation, in contrast to control myotubes. While we observed no differences in the size of intracellular Ca2+ stores, DM1 myotubes showed significantly reduced RyR1 protein levels, uncoupling between the segregated ER/SR Ca2+ store and the voltage-induced Ca2+ release machinery, parallel with induction of endoplasmic reticulum (ER) stress markers. In conclusion, our data suggest that perturbed Ca2+ homeostasis, via activation of ER stress, contributes to muscle degeneration in DM1 muscle cells likely representing a premature senescence phenotype.
- Published
- 2013
48. Local Control of Nuclear Calcium Signaling in Cardiac Myocytes by Perinuclear Microdomains of Sarcolemmal Insulin-Like Growth Factor 1 Receptors
- Author
-
Jaime García-Prieto, Enrique Jaimovich, Juan Pablo Muñoz, Olle Larsson, Sean M. Davidson, Sergio Lavandero, Mario Chiong, Ivana Bulatovic, Manuel Estrada, Paola Rebellato, Gyorgy Szabadkai, Andrew F. G. Quest, Cristian Ibarra, Karl-Henrik Grinnemo, Per Uhlén, Yingbo Lin, Paola Rocco, and Jose Miguel Vicencio
- Subjects
Adult ,Physiology ,Biology ,Article ,Receptor, IGF Type 1 ,Rats, Sprague-Dawley ,Mice ,chemistry.chemical_compound ,Membrane Microdomains ,Sarcolemma ,Animals ,Humans ,Myocyte ,Myocytes, Cardiac ,Inositol ,Calcium Signaling ,Phosphatidylinositol ,Transcription factor ,Cells, Cultured ,Cell Nucleus ,Endoplasmic reticulum ,Rats ,Cell biology ,Mice, Inbred C57BL ,Cytosol ,Animals, Newborn ,chemistry ,Biochemistry ,Cardiology and Cardiovascular Medicine ,Nuclear localization sequence ,Signal Transduction - Abstract
Rationale: The ability of a cell to independently regulate nuclear and cytosolic Ca 2+ signaling is currently attributed to the differential distribution of inositol 1,4,5-trisphosphate receptor channel isoforms in the nucleoplasmic versus the endoplasmic reticulum. In cardiac myocytes, T-tubules confer the necessary compartmentation of Ca 2+ signals, which allows sarcomere contraction in response to plasma membrane depolarization, but whether there is a similar structure tunneling extracellular stimulation to control nuclear Ca 2+ signals locally has not been explored. Objective: To study the role of perinuclear sarcolemma in selective nuclear Ca 2+ signaling. Methods and Results: We report here that insulin-like growth factor 1 triggers a fast and independent nuclear Ca 2+ signal in neonatal rat cardiac myocytes, human embryonic cardiac myocytes, and adult rat cardiac myocytes. This fast and localized response is achieved by activation of insulin-like growth factor 1 receptor signaling complexes present in perinuclear invaginations of the plasma membrane. The perinuclear insulin-like growth factor 1 receptor pool connects extracellular stimulation to local activation of nuclear Ca 2+ signaling and transcriptional upregulation through the perinuclear hydrolysis of phosphatidylinositol 4,5-biphosphate inositol 1,4,5-trisphosphate production, nuclear Ca 2+ release, and activation of the transcription factor myocyte-enhancing factor 2C. Genetically engineered Ca 2+ buffers—parvalbumin—with cytosolic or nuclear localization demonstrated that the nuclear Ca 2+ handling system is physically and functionally segregated from the cytosolic Ca 2+ signaling machinery. Conclusions: These data reveal the existence of an inositol 1,4,5-trisphosphate–dependent nuclear Ca 2+ toolkit located in direct apposition to the cell surface, which allows the local control of rapid and independent activation of nuclear Ca 2+ signaling in response to an extracellular ligand.
- Published
- 2013
49. Loss-of-function mutations in MICU1 cause a brain and muscle disorder linked to primary alterations in mitochondrial calcium signaling
- Author
-
Marjolein Kriek, Gijs W. E. Santen, Colin A. Johnson, Karen Pysden, Eamonn Sheridan, Matthew E. Hurles, Helen Roper, Anna Raffaello, Subaashini Natarajan, Joanne E. Morgan, Zakia Abdelhamed, Ieke B. Ginjaar, Michael R. Duchen, Johan T. den Dunnen, Rosario Rizzuto, Nicola Roberts, Gabrielle Wheway, Katarzyna Szymanska, Diego De Stefani, A. Reghan Foley, Silvia Torelli, Erik H. Niks, Francesco Muntoni, Yu Sun, David A. Parry, Tamieka Whyte, Dick Lindhout, Iulia Munteanu, David T. Bonthron, Clare V. Logan, Caroline Sewry, Jenny Sharpe, Rahul Phadke, Gyorgy Szabadkai, Anne-Marie Childs, and W. Ludo van der Pol
- Subjects
DNA, Complementary ,Molecular Sequence Data ,Fluorescent Antibody Technique ,Mitochondrion ,Muscle disorder ,Biology ,Real-Time Polymerase Chain Reaction ,Mitochondrial Membrane Transport Proteins ,Polymorphism, Single Nucleotide ,Quadriceps Muscle ,Mitochondrial membrane transport protein ,Muscular Diseases ,Learning Disorders ,Calcium-binding protein ,Genetics ,UK10K Consortium ,Humans ,Exome ,Calcium Signaling ,Uniporter ,Inner mitochondrial membrane ,Cation Transport Proteins ,Calcium signaling ,Extrapyramidal Tracts ,Membrane Potential, Mitochondrial ,Analysis of Variance ,Movement Disorders ,Base Sequence ,Learning Disabilities ,Calcium-Binding Proteins ,Histological Techniques ,Sequence Analysis, DNA ,Molecular biology ,Immunohistochemistry ,Cell biology ,Mitochondria ,Pedigree ,Phenotype ,biology.protein ,ATP–ADP translocase ,Calcium Channels - Abstract
Mitochondrial Ca(2+) uptake has key roles in cell life and death. Physiological Ca(2+) signaling regulates aerobic metabolism, whereas pathological Ca(2+) overload triggers cell death. Mitochondrial Ca(2+) uptake is mediated by the Ca(2+) uniporter complex in the inner mitochondrial membrane, which comprises MCU, a Ca(2+)-selective ion channel, and its regulator, MICU1. Here we report mutations of MICU1 in individuals with a disease phenotype characterized by proximal myopathy, learning difficulties and a progressive extrapyramidal movement disorder. In fibroblasts from subjects with MICU1 mutations, agonist-induced mitochondrial Ca(2+) uptake at low cytosolic Ca(2+) concentrations was increased, and cytosolic Ca(2+) signals were reduced. Although resting mitochondrial membrane potential was unchanged in MICU1-deficient cells, the mitochondrial network was severely fragmented. Whereas the pathophysiology of muscular dystrophy and the core myopathies involves abnormal mitochondrial Ca(2+) handling, the phenotype associated with MICU1 deficiency is caused by a primary defect in mitochondrial Ca(2+) signaling, demonstrating the crucial role of mitochondrial Ca(2+) uptake in humans.
- Published
- 2013
- Full Text
- View/download PDF
50. Identification of ER-000444793, a Cyclophilin D-independent inhibitor of mitochondrial permeability transition, using a high-throughput screen in cryopreserved mitochondria
- Author
-
Michael R. Duchen, Malcolm Roberts, Sian Lewis, Yongchun Shen, Hironori Fukumoto, Ben Powney, Naomi Hartopp, Gyorgy Szabadkai, James M. Staddon, Mumta Koglin, Kavita Mistry, Thomas Briston, and Ryosuke Katsumata
- Subjects
0301 basic medicine ,High-throughput screening ,Mitochondrion ,Biology ,Pharmacology ,Mitochondrial Membrane Transport Proteins ,Article ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Cyclophilins ,Adenosine Triphosphate ,Animals ,Humans ,Membrane potential ,chemistry.chemical_classification ,Cryopreservation ,Multidisciplinary ,ATP synthase ,Drug discovery ,Mitochondrial Permeability Transition Pore ,MPTP ,Cell biology ,High-Throughput Screening Assays ,Mitochondria ,Rats ,030104 developmental biology ,Enzyme ,chemistry ,Mitochondrial permeability transition pore ,biology.protein ,Quinolines ,Female ,Energy Metabolism ,Cyclophilin D ,HeLa Cells - Abstract
Growing evidence suggests persistent mitochondrial permeability transition pore (mPTP) opening is a key pathophysiological event in cell death underlying a variety of diseases. While it has long been clear the mPTP is a druggable target, current agents are limited by off-target effects and low therapeutic efficacy. Therefore identification and development of novel inhibitors is necessary. To rapidly screen large compound libraries for novel mPTP modulators, a method was exploited to cryopreserve large batches of functionally active mitochondria from cells and tissues. The cryopreserved mitochondria maintained respiratory coupling and ATP synthesis, Ca2+ uptake and transmembrane potential. A high-throughput screen (HTS), using an assay of Ca2+-induced mitochondrial swelling in the cryopreserved mitochondria identified ER-000444793, a potent inhibitor of mPTP opening. Further evaluation using assays of Ca2+-induced membrane depolarisation and Ca2+ retention capacity also indicated that ER-000444793 acted as an inhibitor of the mPTP. ER-000444793 neither affected cyclophilin D (CypD) enzymatic activity, nor displaced of CsA from CypD protein, suggesting a mechanism independent of CypD inhibition. Here we identified a novel, CypD-independent inhibitor of the mPTP. The screening approach and compound described provides a workflow and additional tool to aid the search for novel mPTP modulators and to help understand its molecular nature.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.