71 results on '"Atan Gross"'
Search Results
2. PERPHERAL BLOOD CELL MITOCHONDRIAL DYSFUNCTION IN MYELODYSPLASTIC SYNDROMECAN BE IMPROVED BY A COMBINATION OF COENZYME Q10 AND CARNITINE
- Author
-
Kalman Filanovsky, Michal Haran, Vita Mirkin, Andrei Braester, Olga Shevetz, Anfisa Stanevsky, Erica Sigler, Ekaterina Votinov, Yehudit Zalzman-Amir, Alain Berrebi, Atan Gross, and Lev Shvidel
- Subjects
myelodysplastic syndrome, mitochondria, oxidative phosphorylation, coenzyme Q10, seahorse XF analyzer. ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Structural mitochondrial abnormalities as well as genetic aberrations in mitochondrial proteins have been known in Myelodysplastic syndrome (MDS) , yet there is currently little data regarding the metabolic properties and energy production of MDS cells. In the current study we used state-of-the-art methods to assess OXPHOS in peripheral blood cells obtained from MDS patients and healthy controls We then assessed the effect of food supplements- Coenzyme Q10 and carnitine on mitochondrial function and hematological response .We show here for the first time that in low risk MDS there is a significant impairment of mitochondrial respiration in peripheral blood cells and this can be improved with food supplements. We also show that such myelodysplastic syndrome, mitochondria, oxidative phosphorylation, coenzyme Q10, seahorse XF analyzer. supplements lead to improvement in cytopenia's and quality of life.
- Published
- 2020
- Full Text
- View/download PDF
3. Fas cell surface death receptor controls hepatic lipid metabolism by regulating mitochondrial function
- Author
-
Flurin Item, Stephan Wueest, Vera Lemos, Sokrates Stein, Fabrizio C. Lucchini, Rémy Denzler, Muriel C. Fisser, Tenagne D. Challa, Eija Pirinen, Youngsoo Kim, Silvio Hemmi, Erich Gulbins, Atan Gross, Lorraine A. O’Reilly, Markus Stoffel, Johan Auwerx, and Daniel Konrad
- Subjects
Science - Abstract
Hepatic steatosis is a common disease closely associated with metabolic syndrome and insulin resistance. Here Item et al. show that Fas, a member of the TNF receptor superfamily, contributes to mitochondrial dysfunction, steatosis development, and insulin resistance under high fat diet.
- Published
- 2017
- Full Text
- View/download PDF
4. Loss of Muscle MTCH2 Increases Whole-Body Energy Utilization and Protects from Diet-Induced Obesity
- Author
-
Liat Buzaglo-Azriel, Yael Kuperman, Michael Tsoory, Yehudit Zaltsman, Liat Shachnai, Smadar Levin Zaidman, Elad Bassat, Inbal Michailovici, Alona Sarver, Eldad Tzahor, Michal Haran, Cecile Vernochet, and Atan Gross
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2017
- Full Text
- View/download PDF
5. Competitive Bone-marrow Transplantations
- Author
-
Maria Maryanovich and Atan Gross
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Competitive bone marrow transplantation assay measures multi-lineage reconstitution of hematopoiesis in irradiated transplant recipient mice. Thus this assay is routinely used to determine haematopoietic stem and progenitor cells (HSPCs) functionality in vivo. The principle of the method is to transplant bone marrow donor cells (derived from transgenic mice of choice) on C57BL6 background together with normal competitor bone marrow. In order to distinguish donor from competitor cells upon transplantation, usually competitor mice are congenic and carry the differential B cell antigen originally designated Ly5.1 and CD45.1.A typical competitive bone marrow transplantation experiment will contain two transplantation groups, donor (transgenic mice of choice and their controls) are transplanted in competition with normal competitors and engraftment efficiency is evaluated in both blood and bone marrow.
- Published
- 2014
- Full Text
- View/download PDF
6. Author Reply to Peer Reviews of MTCH2 cooperates with MFN2 and lysophosphatidic acid synthesis to sustain mitochondrial fusion
- Author
-
Andres Goldman, Michael Mullokandov, Yehudit Zaltsman, Limor Regev, Smadar Levin-Zaidman, and Atan Gross
- Published
- 2023
7. MTCH2 cooperates with MFN2 and lysophosphatidic acid synthesis to sustain mitochondrial fusion
- Author
-
Andres Goldman, Michael Mullokandov, Yehudit Zaltsman, Limor Regev, Smadar Zaidman, and Atan Gross
- Abstract
Mitochondrial dynamics is critical to sustain normal mitochondrial function and is linked to the response of cells to stressful conditions. Fusion of the outer mitochondrial membrane (OMM) is regulated by mitofusin 1 (MFN1) and 2 (MFN2), yet the differential contribution of each of these proteins to this process is less understood. Mitochondrial carrier homolog 2 (MTCH2) was shown to compensate for MFN2’s loss, however its exact function in mitochondrial fusion remains poorly understood. Here we determined the mitochondrial fusion-interplay between MFN1, MFN2 and MTCH2 and demonstrate that MFN2 and MTCH2 play separate, but redundant, roles required for mitochondrial fusion. Loss of either MFN2 or MTCH2 elicits mitochondrial fragmentation that retains mitochondrial plasticity, while loss of both proteins completely impairs mitochondrial fusion. We also show that expression of an MFN2 mutant targeted to the endoplasmic reticulum (ER) is sufficient to restore mitochondrial elongation in MTCH2 KO cells and that this restoration depends on the synthesis of the pro-mitochondrial fusion lipid lysophosphatidic acid (LPA). Moreover, silencing of MFN2 or inhibition of de novo LPA synthesis, revealed the requirement of MTCH2 to sustain mitochondrial plasticity in response to stress. Thus, we unmask two cooperative mechanisms that sustain mitochondrial fusion: one in the OMM, dependent on MTCH2 and MFN1, and independent of MFN2; and a second mechanism in the ER that relies on MFN2 and LPA synthesis.
- Published
- 2022
8. Kynurenic acid, a key L-tryptophan-derived metabolite, protects the heart from an ischemic damage
- Author
-
Einat Bigelman, Metsada Pasmanik-Chor, Bareket Dassa, Maxim Itkin, Sergey Malitsky, Orly Dorot, Edward Pichinuk, Smadar Levin-Zaidman, Nili Dezorella, Atan Gross, Anastasia Abashidze, Yuval Kleinberg, Gad Keren, and Michal Entin-Meer
- Abstract
BackgroundRenal injury induces major changes in plasma and cardiac metabolites. We sought to identify a key metabolite that may affect cardiac mitochondria following an acute kidney injury (AKI) that may be harnessed to protect the heart following an acute ischemic event.Methods and ResultsMetabolomics profiling of cardiac lysates and plasma samples derived from rats that underwent AKI 1 or 7 days earlier by 5/6 nephrectomy versus sham-operated controls was performed. We detected only 26 differential metabolites in both heart and plasma samples at the two selected time points, relative to sham. Out of which, kynurenic acid (kynurenate, KYNA) seemed most relevant. Interestingly, KYNA given at 10 mM concentration significantly rescued the viability of H9C2 cardiac myoblast cells grown under anoxic conditions and largely improved their mitochondrial structure and function as determined by flow cytometry and cell staining with MitoTracker dyes. Moreover, KYNA diluted in the drinking water of animals induced with an acute myocardial infarction, highly enhanced their cardiac recovery according to echocardiography and histopathology.Conclusion and translational aspectKYNA may represent a key metabolite absorbed by the heart following AKI. This metabolite can enhance cardiac cell viability following an ischemic event in a mechanism that is mediated, at least in part, by the protection of the cardiac mitochondria. A short-term administration of KYNA may be highly beneficial in the treatment of the acute phase of kidney disease in order to attenuate progression to CRS and in ischemic cardiac conditions to reduce ischemic myocardial damage.HighlightsThe levels of the L-Tryptophan-derived metabolite, Kynurenic acid (KYNA), are significantly elevated in the heart and the plasma of animals induced with an acute kidney disease.KYNA rescues the viability of cardiac cells from an ischemic damage both in vitro and in vivo.KYNA can protect the structure & function of cardiac mitochondria in H9C2 cardiomyoblast cells upon exposure to anoxia.Graphical abstract
- Published
- 2022
9. Learning Deficits in Adult Mitochondria Carrier Homolog 2 Forebrain Knockout Mouse
- Author
-
Antonella Ruggiero, Atan Gross, Menahem Segal, and Etay Aloni
- Subjects
0301 basic medicine ,Long-Term Potentiation ,Spatial Learning ,Hippocampus ,Mitochondrion ,Biology ,Hippocampal formation ,Mitochondrial Membrane Transport Proteins ,03 medical and health sciences ,Prosencephalon ,medicine ,Animals ,Mitochondrial Carrier Homolog 2 ,Mice, Knockout ,Neurons ,General Neuroscience ,Long-term potentiation ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Motor Skills ,Rotarod Performance Test ,Knockout mouse ,Forebrain ,Female ,Microglia ,Neuron ,Neuroscience - Abstract
Mitochondrial Carrier Homolog 2 (MTCH2) acts as a receptor for the BH3 interacting-domain death agonist (BID) in the mitochondrial outer membrane. Loss of MTCH2 affects mitochondria energy metabolism and function. MTCH2 forebrain conditional KO (MTCH2 BKO) display a deficit in hippocampus-dependent cognitive functions. Here we study age-related MTCH2 BKO behavioral and electrophysiological aspects of hippocampal functions. MTCH2 BKO exhibit impaired spatial but not motor learning and an impairment in long-term potentiation (LTP) in hippocampal slices. Moreover, MTCH2 BKO express an increase in activated microglia, in addition to a reduction in neuron density in the hippocampus, but do not express amyloid-β plaques or neurofibrillary tangles. These results highlight the role of mitochondria in the normal hippocampus-dependent memory formation.
- Published
- 2018
10. MTCH2-mediated mitochondrial fusion drives exit from naïve pluripotency in embryonic stem cells
- Author
-
Michael Mullokandov, Jacob H. Hanna, Dilshad H. Khan, Atan Gross, Emmanuel Amzallag, Yehudit Zaltsman, Amir Bahat, Vladislav Krupalnik, Coral Halperin, Ayelet Erez, Andres Goldman, Alon Silberman, and Aaron D. Schimmer
- Subjects
0301 basic medicine ,Dynamins ,Pluripotent Stem Cells ,Science ,Regulator ,General Physics and Astronomy ,Gene Expression ,Mitochondrion ,Mitochondrial Dynamics ,Mitochondrial Membrane Transport Proteins ,General Biochemistry, Genetics and Molecular Biology ,Article ,GTP Phosphohydrolases ,03 medical and health sciences ,Mice ,Gene expression ,Animals ,lcsh:Science ,Induced pluripotent stem cell ,Cells, Cultured ,Mice, Knockout ,Multidisciplinary ,Microscopy, Confocal ,biology ,Chemistry ,Mouse Embryonic Stem Cells ,General Chemistry ,Nanog Homeobox Protein ,Embryonic stem cell ,Cell biology ,Mitochondria ,Mice, Inbred C57BL ,030104 developmental biology ,Histone ,mitochondrial fusion ,Acetylation ,biology.protein ,lcsh:Q - Abstract
The role of mitochondria dynamics and its molecular regulators remains largely unknown during naïve-to-primed pluripotent cell interconversion. Here we report that mitochondrial MTCH2 is a regulator of mitochondrial fusion, essential for the naïve-to-primed interconversion of murine embryonic stem cells (ESCs). During this interconversion, wild-type ESCs elongate their mitochondria and slightly alter their glutamine utilization. In contrast, MTCH2−/− ESCs fail to elongate their mitochondria and to alter their metabolism, maintaining high levels of histone acetylation and expression of naïve pluripotency markers. Importantly, enforced mitochondria elongation by the pro-fusion protein Mitofusin (MFN) 2 or by a dominant negative form of the pro-fission protein dynamin-related protein (DRP) 1 is sufficient to drive the exit from naïve pluripotency of both MTCH2−/− and wild-type ESCs. Taken together, our data indicate that mitochondria elongation, governed by MTCH2, plays a critical role and constitutes an early driving force in the naïve-to-primed pluripotency interconversion of murine ESCs., Reprogramming of mitochondria metabolism occurs during naïve to primed pluripotency differentiation in mouse embryonic stem cells (ESCs). Here the authors show that mitochondrial MTCH2 regulates mitochondrial fusion and that this fusion is required for naïve to primed pluripotency conversion
- Published
- 2018
11. Fas cell surface death receptor controls hepatic lipid metabolism by regulating mitochondrial function
- Author
-
Fabrizio C. Lucchini, Johan Auwerx, Rémy Denzler, Tenagne D. Challa, Eija Pirinen, Daniel Konrad, Stephan Wueest, Atan Gross, Lorraine A. O'Reilly, Youngsoo Kim, Silvio Hemmi, Erich Gulbins, Sokrates Stein, Flurin Item, Muriel C. Fisser, Markus Stoffel, Vera Lemos, A.I. Virtanen -instituutti, University of Zurich, and Konrad, Daniel
- Subjects
0301 basic medicine ,Male ,Medizin ,General Physics and Astronomy ,Mice, Obese ,Mitochondria, Liver ,Mitochondrion ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,Nonalcoholic fatty liver disease ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Fatty liver ,Fatty Acids ,Type 2 diabetes ,Fas receptor ,Metabolic syndrome ,10124 Institute of Molecular Life Sciences ,3100 General Physics and Astronomy ,Liver ,030220 oncology & carcinogenesis ,medicine.medical_specialty ,Fas Ligand Protein ,Science ,10071 Functional Genomics Center Zurich ,1600 General Chemistry ,Mice, Transgenic ,Biology ,Diet, High-Fat ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Insulin resistance ,Lipid oxidation ,1300 General Biochemistry, Genetics and Molecular Biology ,Internal medicine ,medicine ,Animals ,fas Receptor ,Triglycerides ,General Chemistry ,medicine.disease ,Lipid Metabolism ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,10036 Medical Clinic ,570 Life sciences ,biology ,lcsh:Q ,Steatosis ,Insulin Resistance ,Non-alcoholic fatty liver disease - Abstract
Nonalcoholic fatty liver disease is one of the most prevalent metabolic disorders and it tightly associates with obesity, type 2 diabetes, and cardiovascular disease. Reduced mitochondrial lipid oxidation contributes to hepatic fatty acid accumulation. Here, we show that the Fas cell surface death receptor (Fas/CD95/Apo-1) regulates hepatic mitochondrial metabolism. Hepatic Fas overexpression in chow-fed mice compromises fatty acid oxidation, mitochondrial respiration, and the abundance of mitochondrial respiratory complexes promoting hepatic lipid accumulation and insulin resistance. In line, hepatocyte-specific ablation of Fas improves mitochondrial function and ameliorates high-fat-diet-induced hepatic steatosis, glucose tolerance, and insulin resistance. Mechanistically, Fas impairs fatty acid oxidation via the BH3 interacting-domain death agonist (BID). Mice with genetic or pharmacological inhibition of BID are protected from Fas-mediated impairment of mitochondrial oxidation and hepatic steatosis. We suggest Fas as a potential novel therapeutic target to treat obesity-associated fatty liver and insulin resistance., published version, peerReviewed
- Published
- 2017
12. MTCH2 is a conserved regulator of lipid homeostasis
- Author
-
Yehudit Zaltsman, Siu Sylvia Lee, Michael Platov, Sergiy Libert, Atan Gross, Antonella Ruggiero, Adam B. Francisco, and Veerle Rottiers
- Subjects
0301 basic medicine ,Genetics ,Gene knockdown ,Nutrition and Dietetics ,Endocrinology, Diabetes and Metabolism ,Mutant ,Regulator ,Medicine (miscellaneous) ,Biology ,biology.organism_classification ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Endocrinology ,RNA interference ,Cell culture ,Mitochondrial Carrier Homolog 2 ,Estrogen receptor alpha ,Caenorhabditis elegans - Abstract
Objective More than one-third of U.S. adults have obesity, causing an alarming increase in obesity-related comorbidities such as type 2 diabetes. The functional role of mitochondrial carrier homolog 2 (MTCH2), a human obesity-associated gene, in lipid homeostasis was investigated in Caenorhabditis elegans, cell culture, and mice. Methods In C. elegans, MTCH2/MTCH-1 was depleted, using RNAi and a genetic mutant, and overexpressed to assess its effect on lipid accumulation. In cells and mice, shRNAs against MTCH2 were used for knockdown and MTCH2 overexpression vectors were used for overexpression to study the role of this gene in fat accumulation. Results MTCH2 knockdown reduced lipid accumulation in adipocyte-like cells in vitro and in C. elegans and mice in vivo. MTCH2 overexpression increased fat accumulation in cell culture, C. elegans, and mice. Acute MTCH2 inhibition reduced fat accumulation in animals subjected to a high-fat diet. Finally, MTCH2 influenced estrogen receptor 1 (ESR1) activity. Conclusions MTCH2 is a conserved regulator of lipid homeostasis. MTCH2 was found to be both required and sufficient for lipid homeostasis shifts, suggesting that pharmacological inhibition of MTCH2 could be therapeutic for treatment of obesity and related disorders. MTCH2 could influence lipid homeostasis through inhibition of ESR1 activity.
- Published
- 2017
13. The Mitochondrial Transacylase, Tafazzin, Regulates AML Stemness by Modulating Intracellular Levels of Phospholipids
- Author
-
Mathieu Lupien, Yulia Jitkova, Michael Mullokandov, Helen Loo Yau, Rima Al-awar, James R. Hawley, Rose Hurren, Troy Ketela, S. Kim, Veronique Voisin, Neil MacLean, Daniel D. De Carvalho, Mark D. Minden, Geethu E. Thomas, Val A. Fajardo, Ahmed Aman, Zhenyue Hao, Zaza Khuchua, G. Wei Xu, Gary D. Bader, Richard P. Bazinet, Juan J. Aristizabal Henao, Mingjing Xu, Paul J. LeBlanc, Ayesh K. Seneviratne, Steven M. Claypool, Steven M. Chan, Xiaoming Wang, Atan Gross, Aaron D. Schimmer, Ken D. Stark, David Sharon, Raphaël Chouinard-Watkins, Danny V. Jeyaraju, Caitlin Schafer, and Marcela Gronda
- Subjects
Male ,Cell ,Tafazzin ,Mice, Transgenic ,Mice, SCID ,Mitochondrion ,Biology ,Article ,Mice ,chemistry.chemical_compound ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Cell Line, Tumor ,hemic and lymphatic diseases ,Cardiolipin ,Genetics ,medicine ,Animals ,Humans ,Receptor ,Phospholipids ,030304 developmental biology ,Gene knockdown ,0303 health sciences ,Toll-Like Receptors ,Myeloid leukemia ,Phosphatidylserine ,Cell Biology ,Mitochondria ,Cell biology ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,chemistry ,Doxorubicin ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Female ,Stem cell ,Acyltransferases ,030217 neurology & neurosurgery ,Intracellular ,Signal Transduction ,Transcription Factors - Abstract
Summary Tafazzin (TAZ) is a mitochondrial transacylase that remodels the mitochondrial cardiolipin into its mature form. Through a CRISPR screen, we identified TAZ as necessary for the growth and viability of acute myeloid leukemia (AML) cells. Genetic inhibition of TAZ reduced stemness and increased differentiation of AML cells both in vitro and in vivo. In contrast, knockdown of TAZ did not impair normal hematopoiesis under basal conditions. Mechanistically, inhibition of TAZ decreased levels of cardiolipin but also altered global levels of intracellular phospholipids, including phosphatidylserine, which controlled AML stemness and differentiation by modulating toll-like receptor (TLR) signaling.
- Published
- 2019
14. Mitochondrial carrier homolog 2 is necessary for AML survival
- Author
-
Gary D. Bader, Troy Ketela, Veronique Voisin, Danny V. Jeyaraju, Dilshad H. Khan, G. Wei Xu, Sajid A. Marhon, Yulia Jitkova, Michael Mullokandov, Rob C. Laister, Aaron D. Schimmer, Xiaoming Wang, Rose Hurren, Daniel D. De Carvalho, Marcela Gronda, Ayesh K. Seneviratne, Zachary Blatman, Neil MacLean, Mark D. Minden, Atan Gross, and Yan Wu
- Subjects
0301 basic medicine ,Oncogene Proteins, Fusion ,Cellular differentiation ,Immunology ,Biology ,Mitochondrion ,Biochemistry ,Mitochondrial Membrane Transport Proteins ,Histones ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Cell Line, Tumor ,Pyruvic Acid ,Animals ,Humans ,RNA, Small Interfering ,Mitochondrial Carrier Homolog 2 ,Regulation of gene expression ,Cell Nucleus ,Gene Expression Regulation, Leukemic ,Myeloid leukemia ,Acetylation ,Cell Differentiation ,Cell Biology ,Hematology ,Pyruvate dehydrogenase complex ,Fetal Blood ,Cell biology ,Mitochondria ,Neoplasm Proteins ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,RNA Interference ,Stem cell ,CRISPR-Cas Systems ,Protein Processing, Post-Translational ,Myeloid-Lymphoid Leukemia Protein - Abstract
Through a clustered regularly insterspaced short palindromic repeats (CRISPR) screen to identify mitochondrial genes necessary for the growth of acute myeloid leukemia (AML) cells, we identified the mitochondrial outer membrane protein mitochondrial carrier homolog 2 (MTCH2). In AML, knockdown of MTCH2 decreased growth, reduced engraftment potential of stem cells, and induced differentiation. Inhibiting MTCH2 in AML cells increased nuclear pyruvate and pyruvate dehydrogenase (PDH), which induced histone acetylation and subsequently promoted the differentiation of AML cells. Thus, we have defined a new mechanism by which mitochondria and metabolism regulate AML stem cells and gene expression.
- Published
- 2019
15. PERPHERAL BLOOD CELL MITOCHONDRIAL DYSFUNCTION IN MYELODYSPLASTIC SYNDROMECAN BE IMPROVED BY A COMBINATION OF COENZYME Q10 AND CARNITINE
- Author
-
Erica Sigler, Olga Shevetz, Kalman Filanovsky, Atan Gross, Andrei Braester, Vita Mirkin, Yehudit Zaltsman-Amir, Anfisa Stanevsky, Alain Berrebi, Ekaterina Votinov, Lev Shvidel, and Michal Haran
- Subjects
0301 basic medicine ,Coenzyme Q10 ,Cytopenia ,business.industry ,Hematology ,Oxidative phosphorylation ,Mitochondrion ,Pharmacology ,medicine.disease ,Peripheral blood ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,chemistry ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,Peripheral blood cell ,Carnitine ,business ,Mitochondrial protein ,medicine.drug - Abstract
Structural mitochondrial abnormalities as well as genetic aberrations in mitochondrial proteins have been known in Myelodysplastic syndrome (MDS) , yet there is currently little data regarding the metabolic properties and energy production of MDS cells. In the current study we used state-of-the-art methods to assess OXPHOS in peripheral blood cells obtained from MDS patients and healthy controls We then assessed the effect of food supplements- Coenzyme Q10 and carnitine on mitochondrial function and hematological response .We show here for the first time that in low risk MDS there is a significant impairment of mitochondrial respiration in peripheral blood cells and this can be improved with food supplements. We also show that such myelodysplastic syndrome, mitochondria, oxidative phosphorylation, coenzyme Q10, seahorse XF analyzer. supplements lead to improvement in cytopenia's and quality of life.
- Published
- 2020
16. Obesity, diabetes and zinc: A workshop promoting knowledge and collaboration between the UK and Israel, november 28-30, 2016 - Israel
- Author
-
Arie Moran, Eli C. Lewis, Nicola M Lowe, Amir Tirosh, Guy A. Rutter, Mogher Khamaisi, Wolfgang Maret, Imre Lengyel, Alicia J. Jenkins, Niv Zmora, Atan Gross, Simon C. Andrews, Iris Shai, and Tunde Petro
- Subjects
0301 basic medicine ,Library science ,Clinical epidemiology ,B400 ,Toxicology ,Biochemistry ,Education ,Inorganic Chemistry ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Diabetes Mellitus ,Humans ,Cost action ,Sociology ,Obesity ,Israel ,Related factors ,0601 Biochemistry And Cell Biology ,United Kingdom ,Zinc homeostasis ,Zinc ,030104 developmental biology ,Molecular Medicine - Abstract
Sponsored by the Friends of Israel Educational Foundation (FIEF)\ud and Ben-Gurion University of the Negev and supported by the EU COST\ud action Zinc-Net (COST TD1304), a three-day collaborative UK-Israel\ud workshop was organized by Drs Assaf Rudich, Imre Lengyel and Arie\ud Moran. Participants from the UK and Israel met at the Desert Iris Hotel,\ud Yeruham, Israel between the 28-30th of November 2016 for in-depth\ud discussions, rather than a lecture series, to set the stage for future\ud collaborative grants and projects on diabetes and zinc. Two days of\ud formal scientific sessions with dynamic and wide-ranging discussions\ud was followed by a day of touring and informal networking in the Negev\ud area. This format was previously recognized by our sponsors as both\ud effective and enjoyable and all participants agreed at the end of the\ud meeting that the 3-days provided an excellent basis for future scientific\ud collaboration. The discussions were centered on diabetes and obesity,\ud already at pandemic levels, and zinc homeostasis which is related to the\ud clinical issues and themes of the meeting. The free-flowing discussions\ud were based on short presentations setting the scene for the six main\ud topics: ‘Diabetes and zinc transporters’, ‘Nutrition related factors’,\ud ‘Biomarkers’, ‘Clinical epidemiology’, ‘the Microbiome and diabetes’,\ud and ‘Related diseases’. The abstract style summary of the sessions is\ud followed by the major discussion points raised by the Authors and other\ud participants (UK: Patrik Rorsman, Oxford University; Alan Stewart,\ud University of St Andrews and Israel: Assaf Rudich, Idit Liberty, Rahel\ud Gol, Guy Las and Amos Katz, Ben-Gurion University; Sarah Zangen,\ud Haddassa University). We hope that readers will find this discourse\ud stimulating and some of the ideas might make their way into their research\ud efforts.
- Published
- 2017
17. The IFN-1BIDROS pathway: Linking DNA damage with HSPC malfunction
- Author
-
Alpaslan Tasdogan, Atan Gross, and Hans Joerg Fehling
- Subjects
0301 basic medicine ,Cell type ,Reactive oxygen species metabolism ,DNA damage ,Mitochondrion ,Biology ,Editorials: Cell Cycle Features ,03 medical and health sciences ,Interferon ,medicine ,Animals ,Humans ,Progenitor cell ,Molecular Biology ,Cell Biology ,Hematopoietic Stem Cells ,Haematopoiesis ,030104 developmental biology ,Interferon Type I ,Cancer research ,Signal transduction ,Reactive Oxygen Species ,Developmental Biology ,medicine.drug ,BH3 Interacting Domain Death Agonist Protein ,DNA Damage ,Signal Transduction - Abstract
Haematopoietic stem and progenitor cells (HSPCs), which sustain lifelong production of all haematopoietic cell types, are known to be exquisitely sensitive to DNA-damaging events, like γ-irradiatio...
- Published
- 2017
18. Balancing glycolysis and mitochondrial OXPHOS: Lessons from the hematopoietic system and exercising muscles
- Author
-
Atan Gross and Michal Haran
- Subjects
Hematopoietic System ,Context (language use) ,Cell Biology ,Oxidative phosphorylation ,Biology ,Mitochondrion ,Oxidative Phosphorylation ,Mitochondria ,Cell biology ,Cytosol ,Biochemistry ,Animals ,Molecular Medicine ,Glycolysis ,Energy Metabolism ,Muscle, Skeletal ,Energy source ,Molecular Biology ,Organism ,Homeostasis - Abstract
Living organisms require a constant supply of safe and efficient energy to maintain homeostasis and to allow locomotion of single cells, tissues and the entire organism. The source of energy can be glycolysis, a simple series of enzymatic reactions in the cytosol, or a much more complex process in the mitochondria, oxidative phosphorylation (OXPHOS). In this review we will examine how does the organism balance its source of energy in two seemingly distinct and unrelated processes: hematopoiesis and exercise. In both processes we will show the importance of the metabolic program and its regulation. We will also discuss the importance of oxygen availability not as a sole determinant, but in the context of the nutrient and cellular state, and address the emerging role of lactate as an energy source and signaling molecule in health and disease.
- Published
- 2014
19. Non-apoptotic functions of BCL-2 family proteins
- Author
-
Atan Gross and Samuel G. Katz
- Subjects
0301 basic medicine ,Programmed cell death ,DNA damage ,Cell Survival ,Apoptosis ,Review ,Biology ,Endoplasmic Reticulum ,Electron Transport ,03 medical and health sciences ,Autophagy ,Animals ,Humans ,Molecular Biology ,Regulation of gene expression ,Endoplasmic reticulum ,Bcl-2 family ,Cell Cycle ,Mitophagy ,Cell Biology ,Cell biology ,Mitochondria ,030104 developmental biology ,Glucose ,Gene Expression Regulation ,Proto-Oncogene Proteins c-bcl-2 ,Unfolded protein response ,Unfolded Protein Response ,Calcium ,Signal transduction ,Signal Transduction - Abstract
The BCL-2 family proteins are major regulators of the apoptosis process, but the mechanisms by which they regulate this process are only partially understood. It is now well documented that these proteins play additional non-apoptotic roles that are likely to be related to their apoptotic roles and to provide important clues to cracking their mechanisms of action. It seems that these non-apoptotic roles are largely related to the activation of cellular survival pathways designated to maintain or regain cellular survival, but, if unsuccessful, will switch over into a pro-apoptotic mode. These non-apoptotic roles span a wide range of processes that include the regulation of mitochondrial physiology (metabolism, electron transport chain, morphology, permeability transition), endoplasmic reticulum physiology (calcium homeostasis, unfolded protein response (UPR)), nuclear processes (cell cycle, DNA damage response (DDR)), whole-cell metabolism (glucose and lipid), and autophagy. Here we review all these different non-apoptotic roles, make an attempt to link them to the apoptotic roles, and present many open questions for future research directions in this fascinating field.
- Published
- 2016
20. Loss of forebrain MTCH2 decreases mitochondria motility and calcium handling and impairs hippocampal-dependent cognitive functions
- Author
-
Eduard Korkotian, Efrat Oni-Biton, Ori Brenner, Atan Gross, Smadar Levin-Zaidman, Antonella Ruggiero, Yehudit Zaltsman, Michael Tsoory, Etay Aloni, Yael Kuperman, Liat Shachnai, and Menahem Segal
- Subjects
0301 basic medicine ,Male ,Calcium buffering ,Long-Term Potentiation ,Motility ,Hippocampal formation ,Mitochondrion ,Hippocampus ,Mitochondrial Membrane Transport Proteins ,Synaptic Transmission ,Rotarod performance test ,Article ,03 medical and health sciences ,Mice ,Cognition ,Prosencephalon ,Animals ,Maze Learning ,Postural Balance ,Spatial Memory ,Mice, Knockout ,Neurons ,Multidisciplinary ,Chemistry ,Long-term potentiation ,Anatomy ,Cell biology ,Mitochondria ,030104 developmental biology ,nervous system ,Rotarod Performance Test ,Forebrain ,Excitatory postsynaptic potential ,Calcium ,Female ,Psychomotor Disorders ,Energy Metabolism ,Locomotion - Abstract
Mitochondrial Carrier Homolog 2 (MTCH2) is a novel regulator of mitochondria metabolism, which was recently associated with Alzheimer’s disease. Here we demonstrate that deletion of forebrain MTCH2 increases mitochondria and whole-body energy metabolism, increases locomotor activity, but impairs motor coordination and balance. Importantly, mice deficient in forebrain MTCH2 display a deficit in hippocampus-dependent cognitive functions, including spatial memory, long term potentiation (LTP) and rates of spontaneous excitatory synaptic currents. Moreover, MTCH2-deficient hippocampal neurons display a deficit in mitochondria motility and calcium handling. Thus, MTCH2 is a critical player in neuronal cell biology, controlling mitochondria metabolism, motility and calcium buffering to regulate hippocampal-dependent cognitive functions.
- Published
- 2016
21. tBid Undergoes Multiple Conformational Changes at the Membrane Required for Bax Activation
- Author
-
Atan Gross, Scott Bindner, Clinton J. V. Campbell, Aisha Shamas-Din, Yehudit Zaltsman, Weijia Zhu, Brian Leber, David W. Andrews, and Cécile Fradin
- Subjects
Models, Molecular ,Conformational change ,Time Factors ,Protein Conformation ,Apoptosis ,Plasma protein binding ,Mitochondrion ,Mitochondrial Membrane Transport Proteins ,Models, Biological ,Biochemistry ,Permeability ,Cell membrane ,Mice ,Mitochondrial membrane transport protein ,Protein structure ,Bcl-2-associated X protein ,Fluorescence Resonance Energy Transfer ,medicine ,Animals ,Humans ,Molecular Biology ,bcl-2-Associated X Protein ,Mice, Knockout ,Caspase 8 ,biology ,Cell Membrane ,Cell Biology ,Mitochondria ,Cell biology ,Kinetics ,medicine.anatomical_structure ,Liposomes ,Mitochondrial Membranes ,Mutation ,biology.protein ,Bacterial outer membrane ,BH3 Interacting Domain Death Agonist Protein ,HeLa Cells ,Protein Binding - Abstract
Bid is a Bcl-2 family protein that promotes apoptosis by activating Bax and eliciting mitochondrial outer membrane permeabilization (MOMP). Full-length Bid is cleaved in response to apoptotic stimuli into two fragments, p7 and tBid (p15), that are held together by strong hydrophobic interactions until the complex binds to membranes. The detailed mechanism(s) of fragment separation including tBid binding to membranes and release of the p7 fragment to the cytoplasm remain unclear. Using liposomes or isolated mitochondria with fluorescently labeled proteins at physiological concentrations as in vitro models, we report that the two components of the complex quickly separate upon interaction with a membrane. Once tBid binds to the membrane, it undergoes slow structural rearrangements that result in an equilibrium between two major tBid conformations on the membrane. The conformational change of tBid is a prerequisite for interaction with Bax and is, therefore, a novel step that can be modulated to promote or inhibit MOMP. Using automated high-throughput image analysis in cells, we show that down-regulation of Mtch2 causes a significant delay between tBid and Bax relocalization in cells. We propose that by promoting insertion of tBid via a conformational change at the mitochondrial outer membrane, Mtch2 accelerates tBid-mediated Bax activation and MOMP. Thus the interaction of Mtch2 and tBid is a potential target for therapeutic control of Bid initiated cell death.
- Published
- 2013
22. A ROS rheostat for cell fate regulation
- Author
-
Atan Gross and Maria Maryanovich
- Subjects
Mitochondrial ROS ,Cell ,Apoptosis ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,Cell fate determination ,Mitochondrion ,medicine ,Humans ,Regulation of gene expression ,Tumor Suppressor Proteins ,Cell Cycle ,NF-kappa B ,Cell Biology ,Cell cycle ,Hematopoietic Stem Cells ,Hematopoiesis ,Mitochondria ,Cell biology ,DNA-Binding Proteins ,medicine.anatomical_structure ,Gene Expression Regulation ,Proto-Oncogene Proteins c-bcl-2 ,Tumor Suppressor Protein p53 ,Signal transduction ,Reactive Oxygen Species ,Signal Transduction - Abstract
Cellular reactive oxygen species (ROS) are tightly regulated to prevent tissue damage. ROS also help to monitor different cell fates, suggesting that a 'ROS rheostat' exists in cells. It is well established that ROS are crucial for stem cell biology; in this review, we discuss how mitochondrial ROS influence hematopoietic cell fates. We also examine the importance in this process of BID and other BCL-2 family members, many of which have been implicated in regulating cell fates by modulating mitochondrial integrity/activity and cell cycle progression in the hematopoietic lineage. Based on this knowledge, we propose that selected BCL-2 proteins coordinate mitochondria and nuclear activities via ROS to enable 'synchronized' cell fate decisions.
- Published
- 2013
23. Rejuvenating Bi(d)ology
- Author
-
Sandra S. Zinkel, Atan Gross, and Xiao Ming Yin
- Subjects
Cancer Research ,biology ,Kinase ,DNA damage ,Apoptosis ,Mitochondrion ,Hematopoietic Stem Cells ,medicine.disease ,Mitochondrial carrier ,Article ,Cell biology ,Ataxia-telangiectasia ,Genetics ,biology.protein ,medicine ,Animals ,Humans ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Molecular Biology ,Reperfusion injury ,Caspase ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
The BH3-only Bid protein is a critical sentinel of cellular stress in the liver and the hematopoietic system. Bid's initial `claim to fame' came from its ability—as a caspase-truncated product—to trigger the mitochondrial apoptotic program following death receptor activation. Today we know that Bid can response to multiple types of proteases, which are activated under different conditions such as T-cell activation, ischemical reperfusion injury and lysosomal injury. Activation of the mitochondrial apoptotic program by Bid—via its recently identified receptor mitochondrial carrier homolog 2—involves multiple mechanisms, including release of cytochrome c and second mitochondria-derived activator of caspase (Smac), alteration of mitochondrial cristae organization, generation of reactive oxygen species and engagement of the permeability transition pore. Bid is also emerging—in its full-length form—as a pivotal sentinel of DNA damage in the bone marrow regulated by the ataxia telangiectasia mutated (ATM)/ataxia telangiectasia and Rad3-related (ATR) kinases. The ATM/ATR-Bid pathway is critically involved in preserving the quiescence and survival of hematopoietic stem cells both in the absence and presence of external stress, and a large part of this review will be dedicated to recent advances in this area of research.
- Published
- 2012
24. Molecular Basis of the Interaction between Proapoptotic Truncated BID (tBID) Protein and Mitochondrial Carrier Homologue 2 (MTCH2) Protein
- Author
-
Yehudit Zaltsman-Amir, Yana Mostizky, Assaf Friedler, Chen Katz, Neta Kollet, and Atan Gross
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Programmed cell death ,Truncated BID ,Peptide ,Cell Biology ,Mitochondrion ,TBid Protein ,Biology ,Mitochondrial carrier ,Biochemistry ,3. Good health ,Protein–protein interaction ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Binding site ,Molecular Biology ,030304 developmental biology - Abstract
The molecular basis of the interaction between mitochondrial carrier homologue 2 (MTCH2) and truncated BID (tBID) was characterized. These proteins participate in the apoptotic pathway, and the interaction between them may serve as a target for anticancer lead compounds. In response to apoptotic signals, MTCH2 recruits tBID to the mitochondria, where it activates apoptosis. A combination of peptide arrays screening with biochemical and biophysical techniques was used to characterize the mechanism of the interaction between tBID and MTCH2 at the structural and molecular levels. The regions that mediate the interaction between the proteins were identified. The two specific binding sites between the proteins were determined to be tBID residues 59–73 that bind MTCH2 residues 140–161, and tBID residues 111–125 that bind MTCH2 residues 240–290. Peptides derived from tBID residues 111–125 and 59–73 induced cell death in osteosarcoma cells. These peptides may serve as lead compounds for anticancer drugs that act by targeting the tBID-MTCH2 interaction.
- Published
- 2012
25. The ATM–BID pathway regulates quiescence and survival of haematopoietic stem cells
- Author
-
Lidiya Vorobiyov, Tsvee Lapidot, Steffen Jung, Yehudit Zaltsman, Atan Gross, Hagit Niv, Galia Oberkovitz, Ori Brenner, and Maria Maryanovich
- Subjects
Genetically modified mouse ,Cell Survival ,Cell Cycle Proteins ,Chromosomal translocation ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Mitochondrion ,Biology ,urologic and male genital diseases ,Bid Protein ,Mice ,Animals ,heterocyclic compounds ,Phosphorylation ,neoplasms ,Tumor Suppressor Proteins ,Cell Biology ,Hematopoietic Stem Cells ,digestive system diseases ,Cell biology ,DNA-Binding Proteins ,Haematopoiesis ,ComputingMilieux_COMPUTERSANDSOCIETY ,biological phenomena, cell phenomena, and immunity ,Stem cell ,BH3 Interacting Domain Death Agonist Protein ,DNA Damage - Abstract
BID, a BH3-only BCL2 family member, functions in apoptosis as well as the DNA-damage response. Our previous data demonstrated that BID is an ATM effector acting to induce cell-cycle arrest and inhibition of apoptosis following DNA damage. Here we show that ATM-mediated BID phosphorylation plays an unexpected role in maintaining the quiescence of haematopoietic stem cells (HSCs). Loss of BID phosphorylation leads to escape from quiescence of HSCs, resulting in exhaustion of the HSC pool and a marked reduction of HSC repopulating potential in vivo. We also demonstrate that BID phosphorylation plays a role in protecting HSCs from irradiation, and that regulating both quiescence and survival of HSCs depends on BID's ability to regulate oxidative stress. Moreover, loss of BID phosphorylation, ATM knockout or exposing mice to irradiation leads to an increase in mitochondrial BID, which correlates with an increase in mitochondrial oxidative stress. These results show that the ATM-BID pathway serves as a critical checkpoint for coupling HSC homeostasis and the DNA-damage stress response to enable long-term regenerative capacity.
- Published
- 2012
26. Loss of Muscle MTCH2 Increases Whole-Body Energy Utilization and Protects from Diet-Induced Obesity
- Author
-
Atan Gross, Yehudit Zaltsman, Michael Tsoory, Smadar Levin Zaidman, Eldad Tzahor, Yael Kuperman, Elad Bassat, Liat Buzaglo-Azriel, Michal Haran, Cecile Vernochet, Liat Shachnai, Inbal Michailovici, and Alona Sarver
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Gene Expression ,030209 endocrinology & metabolism ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Diet, High-Fat ,Mitochondrial Membrane Transport Proteins ,Oxidative Phosphorylation ,General Biochemistry, Genetics and Molecular Biology ,Energy homeostasis ,Mice ,03 medical and health sciences ,Mitochondrial membrane transport protein ,0302 clinical medicine ,Endurance training ,Physical Conditioning, Animal ,Internal medicine ,medicine ,Hyperinsulinemia ,Animals ,Humans ,Glycolysis ,Obesity ,Muscle, Skeletal ,Mitochondrial Carrier Homolog 2 ,lcsh:QH301-705.5 ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,medicine.disease ,Mitochondria ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Body Composition ,Metabolome ,biology.protein ,Energy Metabolism - Abstract
SummaryMitochondrial carrier homolog 2 (MTCH2) is a repressor of mitochondrial oxidative phosphorylation (OXPHOS), and its locus is associated with increased BMI in humans. Here, we demonstrate that mice deficient in muscle MTCH2 are protected from diet-induced obesity and hyperinsulinemia and that they demonstrate increased energy expenditure. Deletion of muscle MTCH2 also increases mitochondrial OXPHOS and mass, triggers conversion from glycolytic to oxidative fibers, increases capacity for endurance exercise, and increases heart function. Moreover, metabolic profiling of mice deficient in muscle MTCH2 reveals a preference for carbohydrate utilization and an increase in mitochondria and glycolytic flux in muscles. Thus, MTCH2 is a critical player in muscle biology, modulating metabolism and mitochondria mass as well as impacting whole-body energy homeostasis.
- Published
- 2017
27. BID regulates AIF-mediated caspase-independent necroptosis by promoting BAX activation
- Author
-
Cabon L, Laure Delavallée, Bouharrour A, Hans K. Lorenzo, Marie-Noëlle Brunelle-Navas, Santos A. Susin, Atan Gross, Galán-Malo P, and Team 19
- Subjects
Methylnitronitrosoguanidine ,Programmed cell death ,Cell Survival ,Necroptosis ,Poly ADP ribose polymerase ,Calpains ,Apoptosis ,Models, Biological ,BID ,Mice ,Necrosis ,AIF ,03 medical and health sciences ,0302 clinical medicine ,Bcl-2-associated X protein ,Transduction, Genetic ,Proto-Oncogene Proteins ,Animals ,Molecular Biology ,Caspase ,bcl-2-Associated X Protein ,030304 developmental biology ,Original Paper ,0303 health sciences ,Bcl-2-Like Protein 11 ,biology ,Calpain ,Lentivirus ,Apoptosis Inducing Factor ,Membrane Proteins ,Cell Biology ,Fibroblasts ,Embryo, Mammalian ,3. Good health ,Cell biology ,BAX ,Caspases ,030220 oncology & carcinogenesis ,biology.protein ,Apoptosis-inducing factor ,bcl-Associated Death Protein ,Poly(ADP-ribose) Polymerases ,biological phenomena, cell phenomena, and immunity ,Apoptosis Regulatory Proteins ,BH3 Interacting Domain Death Agonist Protein ,DNA Damage - Abstract
Alkylating DNA-damage agents such as N-methyl-N'-nitro-N'-nitrosoguanidine (MNNG) trigger necroptosis, a newly defined form of programmed cell death (PCD) managed by receptor interacting protein kinases. This caspase-independent mode of cell death involves the sequential activation of poly(ADP-ribose) polymerase-1 (PARP-1), calpains, BAX and AIF, which redistributes from mitochondria to the nucleus to promote chromatinolysis. We have previously demonstrated that the BAX-mediated mitochondrial release of AIF is a critical step in MNNG-mediated necroptosis. However, the mechanism regulating BAX activation in this PCD is poorly understood. Employing mouse embryonic knockout cells, we reveal that BID controls BAX activation in AIF-mediated necroptosis. Indeed, BID is a link between calpains and BAX in this mode of cell death. Therefore, even if PARP-1 and calpains are activated after MNNG treatment, BID genetic ablation abolishes both BAX activation and necroptosis. These PCD defects are reversed by reintroducing the BID-wt cDNA into the BID(-/-) cells. We also demonstrate that, after MNNG treatment, BID is directly processed into tBID by calpains. In this way, calpain non-cleavable BID proteins (BID-G70A or BID-Δ68-71) are unable to promote BAX activation and necroptosis. Once processed, tBID localizes in the mitochondria of MNNG-treated cells, where it can facilitate BAX activation and PCD. Altogether, our data reveal that, as in caspase-dependent apoptosis, BH3-only proteins are key regulators of caspase-independent necroptosis.
- Published
- 2011
28. Programmed Necrotic Cell Death Induced by Complement Involves a Bid-Dependent Pathway
- Author
-
Natalie Donin, Taisia Shmushkovich, Atan Gross, Lea Ziporen, and Zvi Fishelson
- Subjects
Small interfering RNA ,Programmed cell death ,Immunology ,Complement Membrane Attack Complex ,CD59 ,urologic and male genital diseases ,Jurkat cells ,Jurkat Cells ,Mice ,Necrosis ,Animals ,Humans ,Immunology and Allergy ,heterocyclic compounds ,Cytotoxicity ,neoplasms ,Cells, Cultured ,Caspase ,Cell Line, Transformed ,Mice, Knockout ,Cell Death ,biology ,Fibroblasts ,Immunity, Innate ,digestive system diseases ,Cell biology ,Complement system ,Mice, Inbred C57BL ,Animals, Newborn ,Caspases ,biology.protein ,biological phenomena, cell phenomena, and immunity ,K562 Cells ,Complement membrane attack complex ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
The membrane attack complex (MAC) of the complement system induces a necrotic-type cell death. Earlier findings suggested that Bcl-2 protects cells from MAC-induced necrosis. Here we examined the involvement of Bid, a proapoptotic protein, in MAC-induced cytotoxicity. Bid knockout (Bid−/−) mouse embryonic fibroblasts (MEF) and primary fibroblasts were damaged by complement but to a significantly lower extent than wild-type (WT) fibroblasts. Bid silencing with small interfering RNA duplexes led to elevated resistance of mouse fibroblasts, human K562, and Jurkat cells to lysis by complement. Bid−/− MEF were also resistant to toxic doses of streptolysin O, melittin, and A23187. Analysis of complement protein deposition on fibroblasts demonstrated that less complement C3 and C9 bound to Bid−/− than to WT cells, even though expression of the membrane complement inhibitors Crry and CD59 was relatively reduced on Bid−/− cells. Bid was rapidly cleaved in WT MEF subjected to lytic doses of MAC. Pretreatment of the cells with the pan-caspase inhibitor z-Val-Ala-Asp(OMe)-fluoromethylketone reduced Bid cleavage and cell lysis. These results indicate that complement MAC activates two cell death pathways, one involving caspases and Bid and one that is Bid-independent.
- Published
- 2009
29. Nucleocytoplasmic shuttling of BID is involved in regulating its activities in the DNA-damage response
- Author
-
Galia Oberkovitz, Atan Gross, and Limor Regev
- Subjects
Programmed cell death ,DNA damage ,Recombinant Fusion Proteins ,Nuclear Localization Signals ,Active Transport, Cell Nucleus ,Biology ,urologic and male genital diseases ,Cell Line ,S Phase ,Mice ,medicine ,Animals ,Humans ,NLS ,heterocyclic compounds ,Nuclear protein ,Nuclear export signal ,neoplasms ,Molecular Biology ,Etoposide ,Cell Nucleus ,Microscopy, Confocal ,Cell Death ,Cell Biology ,Cell cycle ,Virology ,digestive system diseases ,Cell biology ,Cell nucleus ,medicine.anatomical_structure ,Caspases ,biological phenomena, cell phenomena, and immunity ,Nuclear localization sequence ,BH3 Interacting Domain Death Agonist Protein ,DNA Damage ,HeLa Cells - Abstract
The BH3-only BID protein acts as a sentinel to interconnect specific death signals to the core apoptotic pathway. Our previous data demonstrated that BID is important for both S-phase arrest and cell death following DNA damage, and that the cell cycle arrest function is regulated by its phosphorylation by the ATM kinase. We also showed that a portion of cellular BID localizes to the nucleus. Here, we demonstrate that etoposide and ionizing radiation induce the exit of BID from the nucleus and that leptomycin B, a specific inhibitor of the nuclear export receptor CRM1, prevents the nuclear exit of BID. BID carries a nuclear export signal (NES) consensus motif; however, it does not seem to be functional. To examine the importance of BID nuclear export, we targeted BID to the nucleus by fusing it to a strong nuclear localization signal (NLS). NLS-BID is phosphorylated in a similar time course as wild-type BID, but does not exit the nucleus following etoposide treatment. Importantly, introducing NLS-BID into BID(-/-) cells failed to restore S-phase arrest and cell death in response to etoposide. These results implicate BID as a nuclear protein and raise the possibility that nucleocytoplasmic shuttling of BID is involved in regulating its activities in the DNA-damage response.
- Published
- 2007
30. An MTCH2 pathway repressing mitochondria metabolism regulates haematopoietic stem cell fate
- Author
-
Tsvee Lapidot, Smadar Levin Zaidman, Yehudit Zaltsman, Ziv Porat, Karin Golan, Maria Maryanovich, Andres Goldman, Liat Shachnai, Antonella Ruggiero, and Atan Gross
- Subjects
Blotting, Western ,General Physics and Astronomy ,Apoptosis ,Oxidative phosphorylation ,Biology ,Mitochondrion ,Mitochondrial Size ,Real-Time Polymerase Chain Reaction ,Mitochondrial Membrane Transport Proteins ,Radiation Tolerance ,General Biochemistry, Genetics and Molecular Biology ,Oxidative Phosphorylation ,Colony-Forming Units Assay ,Mice ,Adenosine Triphosphate ,Microscopy, Electron, Transmission ,Animals ,Glycolysis ,Membrane Potential, Mitochondrial ,Multidisciplinary ,Cell Cycle ,Cell Differentiation ,General Chemistry ,Mitochondrial carrier ,Flow Cytometry ,Hematopoietic Stem Cells ,Cell biology ,Hematopoiesis ,Mitochondria ,Haematopoiesis ,Stem cell ,Reactive Oxygen Species ,BH3 Interacting Domain Death Agonist Protein - Abstract
The metabolic state of stem cells is emerging as an important determinant of their fate. In the bone marrow, haematopoietic stem cell (HSC) entry into cycle, triggered by an increase in intracellular reactive oxygen species (ROS), corresponds to a critical metabolic switch from glycolysis to mitochondrial oxidative phosphorylation (OXPHOS). Here we show that loss of mitochondrial carrier homologue 2 (MTCH2) increases mitochondrial OXPHOS, triggering HSC and progenitor entry into cycle. Elevated OXPHOS is accompanied by an increase in mitochondrial size, increase in ATP and ROS levels, and protection from irradiation-induced apoptosis. In contrast, a phosphorylation-deficient mutant of BID, MTCH2's ligand, induces a similar increase in OXPHOS, but with higher ROS and reduced ATP levels, and is associated with hypersensitivity to irradiation. Thus, our results demonstrate that MTCH2 is a negative regulator of mitochondrial OXPHOS downstream of BID, indispensible in maintaining HSC homeostasis.
- Published
- 2015
31. BCL2 family in DNA damage and cell cycle control
- Author
-
E Yang, Atan Gross, and Sandra S. Zinkel
- Subjects
Cell cycle checkpoint ,DNA damage ,Bcl-xL ,medicine.disease_cause ,hemic and lymphatic diseases ,Puma ,medicine ,Animals ,Humans ,CHEK1 ,neoplasms ,Molecular Biology ,biology ,Cell Cycle ,Cell Biology ,Cell cycle ,biology.organism_classification ,Cell biology ,Cell Transformation, Neoplastic ,Proto-Oncogene Proteins c-bcl-2 ,Apoptosis ,biology.protein ,Tumor Suppressor Protein p53 ,biological phenomena, cell phenomena, and immunity ,Carcinogenesis ,DNA Damage - Abstract
Individual BCL2 family members couple apoptosis regulation and cell cycle control in unique ways. Antiapoptotic BCL2 and BCL-x(L) are antiproliferative by facilitating G0. BAX is proapoptotic and accelerates S-phase progression. The dual functions in apoptosis and cell cycle are coordinately regulated by the multi-domain BCL2 family members (MCL-1) and suggest that survival is maintained at the expense of proliferation. The role of BH3-only molecules in cell cycle is more variable. BAD antagonizes both the cell cycle and antiapoptotic functions of BCL2 and BCL-x(L) through BH3 binding. BID has biochemically separable functions in apoptosis and S-phase checkpoint, determined by post-translational modification. p53-induced PUMA is known only to have apoptotic function. Inhibition of apoptosis is oncogenic, whereas promotion of cell cycle arrest is tumor suppressive. Paradoxically, selected BCL2 family members can be both oncogenic and tumor suppressive. Which of the dual functions predominates is lineage specific and context dependent.
- Published
- 2006
32. The BH3-only protein BID impairs the p38-mediated stress response and promotes hepatocarcinogenesis during chronic liver injury in mice
- Author
-
Benjamin Goeppert, Johanna Orlik, Atan Gross, S Rössler, Silke Marhenke, Jessica Endig, Arndt Vogel, Robert Geffers, Michael P. Manns, Laura Elisa Buitrago-Molina, Katharina Lobschat, Sven Schüngel, and Hannover Medical School, Hannover, Germany.
- Subjects
medicine.medical_specialty ,DNA damage ,Carcinogenesis ,Cell Survival ,MAP Kinase Signaling System ,Apoptosis ,Biology ,medicine.disease_cause ,Malignant transformation ,Mice ,Random Allocation ,Liver Neoplasms, Experimental ,Reference Values ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Hepatic Insufficiency ,Tissue homeostasis ,Cell Proliferation ,Liver injury ,Mice, Knockout ,Analysis of Variance ,Hepatology ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Gene Expression Regulation ,Hepatocyte ,Cancer research ,Hepatocytes ,biological phenomena, cell phenomena, and immunity ,Liver cancer ,BH3 Interacting Domain Death Agonist Protein - Abstract
Apoptosis is critical for maintaining tissue homeostasis, and apoptosis evasion is considered as a hallmark of cancer. However, increasing evidence also suggests that proapoptotic molecules can contribute to the development of cancer, including liver cancer. The aim of this study was to further clarify the role of the proapoptotic B-cell lymphoma 2 homology domain 3 (BH3)-only protein BH3 interacting-domain death agonist (BID) for chronic liver injury (CLI) and hepatocarcinogenesis (HCG). Loss of BID significantly delayed tumor development in two mouse models of Fah-mediated and HBsTg-driven HCG, suggesting a tumor-promoting effect of BID. Liver injury as well as basal and mitogen-stimulated hepatocyte proliferation were not modulated by BID. Moreover, there was no in vivo or in vitro evidence that BID was involved in DNA damage response in hepatocytes and hepatoma cells. Our data revealed that CLI was associated with strong activation of oxidative stress (OS) response and that BID impaired full activation of p38 after OS. Conclusion: We provide evidence that the tumor-promoting function of BID in CLI is not related to enhanced proliferation or an impaired DNA damage response. In contrast, BID suppresses p38 activity and facilitates malignant transformation of hepatocytes. (Hepatology 2015;62:816–828)
- Published
- 2014
33. A Role for Proapoptotic BID in the DNA-Damage Response
- Author
-
Sandra S. Zinkel, Christy C. Ong, Atan Gross, Kristen E. Hurov, Stanley J. Korsmeyer, and Farvardean M. Abtahi
- Subjects
Male ,Myeloid ,Cell ,Apoptosis ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,urologic and male genital diseases ,S Phase ,Mice ,0302 clinical medicine ,heterocyclic compounds ,Phosphorylation ,Cell Line, Transformed ,Mice, Knockout ,Genetics ,0303 health sciences ,Kinase ,Leukemia, Myelomonocytic, Chronic ,Cell biology ,DNA-Binding Proteins ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,biological phenomena, cell phenomena, and immunity ,BH3 Interacting Domain Death Agonist Protein ,DNA damage ,Protein Serine-Threonine Kinases ,Biology ,Genomic Instability ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Animals ,neoplasms ,Cell damage ,Myeloid Progenitor Cells ,030304 developmental biology ,Biochemistry, Genetics and Molecular Biology(all) ,Tumor Suppressor Proteins ,medicine.disease ,digestive system diseases ,Protein Structure, Tertiary ,Genes, cdc ,NIH 3T3 Cells ,Carrier Proteins ,Function (biology) ,DNA Damage ,Mutagens - Abstract
SummaryThe BCL-2 family of apoptotic proteins encompasses key regulators proximal to irreversible cell damage. The BH3-only members of this family act as sentinels, interconnecting specific death signals to the core apoptotic pathway. Our previous data demonstrated a role for BH3-only BID in maintaining myeloid homeostasis and suppressing leukemogenesis. In the absence of Bid, mice accumulate chromosomal aberrations and develop a fatal myeloproliferative disorder resembling chronic myelomonocytic leukemia. Here, we describe a role for BID in preserving genomic integrity that places BID at an early point in the path to determine the fate of a cell. We show that BID plays an unexpected role in the intra-S phase checkpoint downstream of DNA damage distinct from its proapoptotic function. We further demonstrate that this role is mediated through BID phosphorylation by the DNA-damage kinase ATM. These results establish a link between proapoptotic Bid and the DNA-damage response.
- Published
- 2005
34. Proapoptotic BID Is an ATM Effector in the DNA-Damage Response
- Author
-
Rachel Sarig, Iris Kamer, Yehudit Zaltsman, Richard C. Marcellus, Limor Regev, Yaniv Lerenthal, Gal Haimovich, Hagit Niv, Atan Gross, and Galia Oberkovitz
- Subjects
Cell Survival ,DNA damage ,Apoptosis ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,urologic and male genital diseases ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,S Phase ,Mice ,medicine ,Animals ,Humans ,Topoisomerase II Inhibitors ,heterocyclic compounds ,Phosphorylation ,neoplasms ,Cells, Cultured ,S phase ,Etoposide ,Nucleic Acid Synthesis Inhibitors ,Cell Nucleus ,Mice, Knockout ,Genetics ,Mutation ,Binding Sites ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Effector ,Tumor Suppressor Proteins ,Topoisomerase ,DNA ,Fibroblasts ,digestive system diseases ,Cell biology ,DNA-Binding Proteins ,Genes, cdc ,Mice, Inbred C57BL ,DNA Topoisomerases, Type II ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Topoisomerase-II Inhibitor ,Carrier Proteins ,BH3 Interacting Domain Death Agonist Protein ,DNA Damage - Abstract
SummaryThe “BH3-only” proapoptotic BCL-2 family members are sentinels of intracellular damage. Here, we demonstrated that the BH3-only BID protein partially localizes to the nucleus in healthy cells, is important for apoptosis induced by DNA damage, and is phosphorylated following induction of double-strand breaks in DNA. We also found that BID phosphorylation is mediated by the ATM kinase and occurs in mouse BID on two ATM consensus sites. Interestingly, BID−/− cells failed to accumulate in the S phase of the cell cycle following treatment with the topoisomerase II poison etoposide; reintroducing wild-type BID restored accumulation. In contrast, introducing a nonphosphorylatable BID mutant did not restore accumulation in the S phase and resulted in an increase in cellular sensitivity to etoposide-induced apoptosis. These results implicate BID as an ATM effector and raise the possibility that proapoptotic BID may also play a prosurvival role important for S phase arrest.
- Published
- 2005
35. Mitochondrial Carrier Homolog 2: A Clue to Cracking the BCL-2 Family Riddle?
- Author
-
Atan Gross
- Subjects
Physiology ,Molecular Sequence Data ,Apoptosis ,Mitochondrion ,Biology ,Mitochondrial Membrane Transport Proteins ,Mitochondrial Proteins ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Mitochondrial Carrier Homolog 2 ,bcl-2-Associated X Protein ,Genetics ,Bcl-2 family ,Membrane Transport Proteins ,Cell Biology ,Mitochondrial carrier ,Genes, bcl-2 ,Mitochondria ,Cell biology ,bcl-2 Homologous Antagonist-Killer Protein ,Proto-Oncogene Proteins c-bcl-2 ,Mechanism of action ,mitochondrial fusion ,DNAJA3 ,medicine.symptom ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
BCL-2 family members are pivotal regulators of the apoptotic process. Mitochondria are a major site-of-action for these proteins. Several prominent alterations occur to mitochondria during apoptosis that seem to be part of the "mitochondrial apoptotic program." The BCL-2 family members are believed to be the major regulators of this program, however their exact mechanism of action still remains a mystery. BID, a pro-apoptotic BCL-2 family member plays an essential role in initiating this program. Recently, we have revealed that in apoptotic cells the activated/truncated form of BID, tBID, interacts with a novel, uncharacterized protein named mitochondrial carrier homolog 2 (Mtch2). Mtch2 is a conserved protein that is similar to members of the mitochondrial carrier protein (MCP) family. This review summarizes the current knowledge regarding BCL-2 family members and the mitochondrial apoptotic program and examines the possible involvement of Mtch2 in this program.
- Published
- 2005
36. Gonadotropins Enhance Caspase-3 and -7 Activity and Apoptosis in the Theca-Interstitial Cells of Rat Preovulatory Follicles in Culture
- Author
-
Alex Tsafriri, Keren Yacobi, Anna Wójtowicz, and Atan Gross
- Subjects
endocrine system ,medicine.medical_specialty ,Time Factors ,medicine.drug_class ,Apoptosis ,Ovary ,Caspase 3 ,DNA Fragmentation ,Endocrinology ,Culture Techniques ,Internal medicine ,Follicular phase ,medicine ,Animals ,Humans ,Rats, Wistar ,Ovarian follicle ,Caspase ,Caspase 7 ,Granulosa Cells ,Sheep ,biology ,Luteinizing Hormone ,Recombinant Proteins ,Rats ,medicine.anatomical_structure ,Follicular Phase ,Theca ,Caspases ,Theca Cells ,biology.protein ,Female ,Follicle Stimulating Hormone ,Gonadotropin - Abstract
Apoptosis causes the elimination of ovarian germ cells and the atretic degeneration of ovarian follicles. Here we have used cultured rat preovulatory follicles to examine the regulation of effector caspase-3 and -7 in follicles undergoing apoptosis in the presence or absence of gonadotropins or IGF-I. Culturing follicles in the presence or absence of serum resulted in the induction of apoptosis of granulosa cells (GC), which was accompanied by effector caspase activation. Surprisingly, the addition of the survival factors LH or FSH, but not IGF-I, further increased caspase-3 and -7 activity. Immunohistochemistry studies of the LH- and FSH-treated follicles indicated that cleaved caspase-3 was predominantly localized to the peripheral theca-interstitial cells (TIC). Western blot analysis and caspase-3 and -7 activity assays of the separated follicular compartments confirmed that both LH and FSH treatments significantly enhance caspase-3 and -7 activity in TIC. The elevation in caspase-3 and -7 activity in TIC was accompanied by an increase in apoptosis. Interestingly, LH and FSH also induced an increase in caspase-3 and -7 activity in GC; however, this increase was accompanied by a decrease in apoptosis. Finally, we demonstrate that in freshly isolated preovulatory follicles and in antral follicles in intact ovaries, the expression level of procaspase-3 is significantly higher in TIC than in GC. Thus, LH and FSH have a dual effect on the cultured rat preovulatory follicle: an antiapoptotic effect on GC and a proapoptotic effect on TIC.
- Published
- 2004
37. The distribution and apoptotic function of outer membrane proteins depend on mitochondrial fusion
- Author
-
Atan Gross, Péter Várnai, David Weaver, György Hajnóczky, Verónica Eisner, László Hunyady, and Xingguo Liu
- Subjects
Muscle Fibers, Skeletal ,MFN2 ,Mitochondrion ,Biology ,Mitochondrial apoptosis-induced channel ,Mitochondrial Dynamics ,Article ,Cell Line ,GTP Phosphohydrolases ,Mitochondrial Proteins ,Gene Knockout Techniques ,Mice ,MFN1 ,Inner membrane ,Animals ,Humans ,Molecular Biology ,Voltage-Dependent Anion Channel 2 ,Cell Biology ,Mitochondrial carrier ,Cell biology ,Rats ,Protein Transport ,bcl-2 Homologous Antagonist-Killer Protein ,mitochondrial fusion ,Mitochondrial Membranes ,biological phenomena, cell phenomena, and immunity ,Bcl-2 Homologous Antagonist-Killer Protein ,BH3 Interacting Domain Death Agonist Protein - Abstract
Cells deficient in mitochondrial fusion have been shown to have defects linked to the exchange of inner membrane and matrix components. Because outer-mitochondrial membrane (OMM) constituents insert directly from the cytoplasm, a role for fusion in their intermitochondrial transfer was unanticipated. Here, we show that fibroblasts lacking the GTPases responsible for OMM fusion, mitofusins 1 and 2 (MFN1 and MFN2), display more heterogeneous distribution of OMM proteins. Proteins with different modes of OMM association display varying degrees of heterogeneity in Mfn1/2(-/-) cells and different kinetics of transfer during fusion in fusion-competent cells. Proapoptotic Bak exhibits marked heterogeneity, which is normalized upon expression of MFN2. Bak is critical for Bid-induced OMM permeabilization and cytochrome c release, and Mfn1/2(-/-) cells show dysregulation of Bid-dependent apoptotic signaling. Bid sensitivity of Bak-deficient mitochondria is regained upon fusion with Bak-containing mitochondria. Thus, OMM protein distribution depends on mitochondrial fusion and is a locus of apoptotic dysfunction in conditions of fusion deficiency.
- Published
- 2014
38. BCL-2 Proteins: Regulators of the Mitochondrial Apoptotic Program
- Author
-
Atan Gross
- Subjects
Apoptotic program ,Clinical Biochemistry ,Apoptosis ,Mitochondrion ,Biology ,Biochemistry ,Permeability ,Apoptotic Process ,Genetics ,medicine ,Animals ,Homeostasis ,Molecular Biology ,Membrane potential ,Membrane Proteins ,Intracellular Membranes ,Cell Biology ,Mitochondria ,Cell biology ,Proto-Oncogene Proteins c-bcl-2 ,Mechanism of action ,sense organs ,medicine.symptom ,Intermembrane space - Abstract
BCL-2 family members are pivotal regulators of the apoptotic process. Mitochondria seem to be a major site-of-action for these proteins. Several prominent alterations occur to mitochondria during apoptosis that include the release of intermembrane space molecules, changes in the membrane potential, ionic changes, and more. All these changes seem to be part of the mitochondrial apoptotic process. The BCL-2 family members are believed to be the major regulators of this program; however, their exact mechanism of action still remains a mystery. In addition, the exact contribution of mitochondria to the apoptotic process is still unclear. This review summarizes and examines the current knowledge regarding these two issues.
- Published
- 2001
39. The Mitochondrial Carrier Homolog 2 (MTCH2) Regulates the Differentiation of AML Cells By Influencing the Localization of Pyruvate Dehydrogenase Complex and H3 and H4 Histone Acetylation
- Author
-
Michael Mullokandov, Rose Hurren, Dilshad H. Khan, Atan Gross, Xiaoming Wang, Neil MacLean, Aaron D. Schimmer, Yan Wu, Marcela Gronda, and Rob C. Laister
- Subjects
Gene knockdown ,Chemistry ,Cellular differentiation ,Immunology ,Mitochondrial pyruvate dehydrogenase complex ,Cell Biology ,Hematology ,Mitochondrion ,Pyruvate dehydrogenase complex ,Biochemistry ,Cell biology ,Mitochondrial respiratory chain ,Mitochondrial Carrier Homolog 2 ,Mitochondrial DNA replication - Abstract
Mitochondrial carrier homolog 2 (MTCH2) is a mitochondrial outer membrane protein that functions as a receptor-like protein for pro-apoptotic BID. In addition to its role in apoptosis, recent findings show that MTCH2 also regulates cellular metabolism. In murine hematopoietic cells, loss of MTCH2 increases oxidative phosphorylation and reduces the number of hematopoietic stem cells. Here, we sought to understand the role of MTCH2 in leukemogenesis and knocked down MTCH2 in leukemia cell lines using multiple independent shRNAs. Knockdown of MTCH2 reduced growth and viability of AML cells: OCI-AML2 (>90%), TEX (>80%), U937 (>65%), and HL60 (>75%). MTCH2 knockdown also decreased the clonogenic growth of OCI-AML2 (>60%), TEX (>70%), and U937 (>40%) cells compared to controls. However, MTCH2 knockdown did not induce cell death as indicated by annexin V/PI staining. In addition, knockdown of MTCH2 in TEX cells reduced engraftment into the marrow of non-obese diabetic/severe combined immunodeficiency-growth factor (NOD/SCID-GF) mice (control 17±4%, n=10) vs. sh-MTCH2 (4±0.86%, n=10). In mouse models, knockout of MTCH2 decreased the leukomogenic potential of murine hematopoietic stem cells transformed with the MLL-AF9 oncogene and increased the survival of these mice. To understand the mechanism by which MTCH2 knockdown decreased cell growth, we used genome wide transcriptome analysis with RNA-seq and observed an up regulation of genes involved in cellular differentiation. Consistent with increased MTCH2 knockdown promoting differentiation, OCI-AML2 cells with MTCH2 knockdown displayed increased non-specific esterase staining. Increased differentiation (Lin+ve cells) was also observed in MLL-AF9 with MTCH2 knockout. Knockdown of MTCH2 in TEX and OCI-AML2 cells increased levels of H3 and H4 histone acetylation as demonstrated by immunoblotting. Of note, differentiation and increased H3 and H4 acetylation was not observed after inhibiting other mitochondrial processes, such as mitochondrial protein synthesis or mitochondrial DNA replication. Although MTCH2 is a receptor for BID, the increased H3 and H4 acetylation appeared independent of BID as small molecule BID inhibitors did not alter H3 and H4 acetylation. MTCH2 regulates cell metabolism. Therefore, we measured changes in intracellular metabolites in AML cells after MTCH2 knockdown. In AML cells, MTCH2 knockdown increased levels of lactate (2 fold), but did not change the basal rate of oxygen consumption or the activity of mitochondrial respiratory chain complexes. Loss of mitochondrial pyruvate dehydrogenase complex increases lactate levels and a recent study reported that the translocation of pyruvate dehydrogenase complex from the mitochondria to the nucleus under conditions of mitochondrial stress, increases H3 and H4 histone acetylation (Cell. 2014; 158(1):84-97). Therefore, we measured changes in the localization of pyruvate dehydrogenase complex after MTCH2 knockdown. Knockdown of MTCH2 decreased mitochondrial and increased nuclear dehydrogenase complex in OCI-AML2 and TEX cells. Thus, in summary, MTCH2 regulates the differentiation of AML cells and controls the localization of pyruvate dehydrogenase complex and histone acetylation. These results also suggest a mechanism by which loss of MTCH2 leads to reductions of normal hematopoietic stem cells. Disclosures Schimmer: Novartis: Honoraria.
- Published
- 2016
40. tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c
- Author
-
Tullia Lindsten, Craig B. Thompson, Stanley J. Korsmeyer, Vamsi K. Mootha, Solly Weiler, Michael C. Wei, Mona Ashiya, and Atan Gross
- Subjects
BH3 Mimetic ABT-737 ,Cytochrome ,Truncated BID ,Allosteric regulation ,Cytochrome c Group ,Mitochondrion ,Cell membrane ,Mice ,Biopolymers ,Allosteric Regulation ,Genetics ,medicine ,Animals ,biology ,Cytochrome c ,Cell Membrane ,Membrane Proteins ,Cell biology ,bcl-2 Homologous Antagonist-Killer Protein ,medicine.anatomical_structure ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,Bcl-2 Homologous Antagonist-Killer Protein ,Research Paper ,BH3 Interacting Domain Death Agonist Protein ,Developmental Biology - Abstract
TNFR1/Fas engagement results in the cleavage of cytosolic BID to truncated tBID, which translocates to mitochondria. Immunodepletion and gene disruption indicate BID is required for cytochrome c release. Surprisingly, the three-dimensional structure of this BH3 domain-only molecule revealed two hydrophobic α-helices suggesting tBID itself might be a pore-forming protein. Instead, we demonstrate that tBID functions as a membrane-targeted death ligand in which an intact BH3 domain is required for cytochrome c release, but not for targeting.Bak-deficient mitochondria and blocking antibodies reveal tBID binds to its mitochondrial partner BAK to release cytochrome c, a process independent of permeability transition. Activated tBID results in an allosteric activation of BAK, inducing its intramembranous oligomerization into a proposed pore for cytochrome c efflux, integrating the pathway from death receptors to cell demise.
- Published
- 2000
41. Biochemical and Genetic Analysis of the Mitochondrial Response of Yeast to BAX and BCL-XL
- Author
-
Jennifer Jockel, Stanley J. Korsmeyer, Elizabeth Blachly-Dyson, Emy Basso, Atan Gross, Michael Forte, Michael C. Bassik, and Kirsten Pilcher
- Subjects
Time Factors ,Voltage-dependent anion channel ,Blotting, Western ,Genes, Fungal ,bcl-X Protein ,Apoptosis ,Mitochondrion ,Mitochondrial apoptosis-induced channel ,Bcl-2-associated X protein ,Proto-Oncogene Proteins ,Yeasts ,Cell Growth and Development ,Molecular Biology ,bcl-2-Associated X Protein ,biology ,Galactose ,Intracellular Membranes ,Cell Biology ,Flow Cytometry ,Molecular biology ,Mitochondria ,Cell biology ,Glucose ,Cell killing ,Proto-Oncogene Proteins c-bcl-2 ,Mitochondrial permeability transition pore ,Mutation ,biology.protein ,DNAJA3 ,ATP–ADP translocase ,Reactive Oxygen Species ,Cell Division ,Subcellular Fractions - Abstract
The BCL-2 family includes both proapoptotic (e.g., BAX and BAK) and antiapoptotic (e.g., BCL-2 and BCL-X(L)) molecules. The cell death-regulating activity of BCL-2 members appears to depend on their ability to modulate mitochondrial function, which may include regulation of the mitochondrial permeability transition pore (PTP). We examined the function of BAX and BCL-X(L) using genetic and biochemical approaches in budding yeast because studies with yeast suggest that BCL-2 family members act upon highly conserved mitochondrial components. In this study we found that in wild-type yeast, BAX induced hyperpolarization of mitochondria, production of reactive oxygen species, growth arrest, and cell death; however, cytochrome c was not released detectably despite the induction of mitochondrial dysfunction. Coexpression of BCL-X(L) prevented all BAX-mediated responses. We also assessed the function of BCL-X(L) and BAX in the same strain of Saccharomyces cerevisiae with deletions of selected mitochondrial proteins that have been implicated in the function of BCL-2 family members. BAX-induced growth arrest was independent of the tested mitochondrial components, including voltage-dependent anion channel (VDAC), the catalytic beta subunit or the delta subunit of the F(0)F(1)-ATP synthase, mitochondrial cyclophilin, cytochrome c, and proteins encoded by the mitochondrial genome as revealed by [rho(0)] cells. In contrast, actual cell killing was dependent upon select mitochondrial components including the beta subunit of ATP synthase and mitochondrial genome-encoded proteins but not VDAC. The BCL-X(L) protection from either BAX-induced growth arrest or cell killing proved to be independent of mitochondrial components. Thus, BAX induces two cellular processes in yeast which can each be abrogated by BCL-X(L): cell arrest, which does not require aspects of mitochondrial biochemistry, and cell killing, which does.
- Published
- 2000
42. The N-Terminal Half of Cdc25 Is Essential for Processing Glucose Signaling in Saccharomyces cerevisiae
- Author
-
Sabina Winograd, Alexander Levitzki, Irit Marbach, and Atan Gross
- Subjects
GTP' ,G protein ,Cdc25 ,Mutant ,Saccharomyces cerevisiae ,Cell Cycle Proteins ,Biochemistry ,Fungal Proteins ,Catalytic Domain ,Animals ,Humans ,Phosphorylation ,Protein kinase A ,Sequence Deletion ,chemistry.chemical_classification ,biology ,ras-GRF1 ,Chemistry ,biology.organism_classification ,Guanine Nucleotides ,Peptide Fragments ,Cell biology ,Amino acid ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,Glucose ,Guanylate Cyclase ,Mutagenesis, Site-Directed ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Protein Processing, Post-Translational ,Tyrosine kinase ,Signal Transduction - Abstract
Saccharomyces cerevisiae Cdc25 is the prototype Ras GDP/GTP exchange protein. Its C-terminal catalytic domain was found to be highly conserved in the homologues p140(ras-GRF) and Sos. The regulatory domains in each Ras exchanger mediate the signals arriving from upstream elements such as tyrosine kinases for Sos, or Ca2+ and G proteins for p140.(Ras-GRF) In this study, we show that the N-terminal half (NTH) of S. cerevisiae Cdc25, as well as the C-terminal 37 amino acids, is essential for processing the elevation of cAMP in response to glucose. The mammalian p140(ras-GRF) catalytic domain (CGRF) restores glucose signaling in S. cerevisiae only if tethered between the N-terminal half (NTH) of S. cerevisiae Cdc25 and the C-terminal 37 amino acids. The glucose-induced transient elevation in cAMP is nullified or severely hampered by the deletion of domains within the NTH of Cdc25. These deletions, however, do not modify the intrinsic GDP/GTP exchange activity of mutant proteins as compared to native Cdc25. We also show that 7 Ser to Ala mutations at the cAMP-dependent protein kinase putative phosphorylation sites within the NTH of Cdc25 eliminate the descending portion of the glucose response curve, responsible for signal termination. These findings support a dual role of the NTH of Cdc25 in both enabling the glucose signal and being responsible for its attenuation.
- Published
- 1999
43. Bid-deficient mice are resistant to Fas-induced hepatocellular apoptosis
- Author
-
Kun Wang, Kevin A. Roth, Stanley J. Korsmeyer, Barbara J. Klocke, Sandra S. Zinkel, Atan Gross, Yongge Zhao, and Xiao Ming Yin
- Subjects
Male ,Programmed cell death ,Truncated BID ,Apoptosis ,Cytochrome c Group ,Mitochondrion ,Caspase 8 ,Receptors, Tumor Necrosis Factor ,Membrane Potentials ,Mice ,Antigens, CD ,Proto-Oncogene Proteins ,Animals ,fas Receptor ,Cells, Cultured ,Caspase ,Multidisciplinary ,biology ,Cytochrome c ,Fas receptor ,Mitochondria ,Cell biology ,Enzyme Activation ,Mice, Inbred C57BL ,Liver ,Biochemistry ,Receptors, Tumor Necrosis Factor, Type I ,Caspases ,biology.protein ,Carrier Proteins ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
The protein Bid is a participant in the pathway that leads to cell death (apoptosis), mediating the release of cytochrome c from mitochondria in response to signals from 'death' receptors known as TNFR1/Fas on the cell surface. It is a member of the proapoptotic Bcd-2 family and is activated as a result of its cleavage by caspase 8, one of a family of proteolytic cell-death proteins. To investigate the role of Bid in vivo, we have generated mice deficient for Bid. We find that when these mice are injected with an antibody directed against Fas, they nearly all survive, whereas wild-type mice die from hepatocellular apoptosis and haemorrhagic necrosis. About half of the Bid-deficient animals had no apparent liver injury and showed no evidence of activation of the effector caspases 3 and 7, although the initiator caspase 8 had been activated. Other Bid-deficient mice survived with only moderate damage: all three caspases (8 and 37) were activated but their cell nuclei were intact and no mitochondrial cytochrome c was released. We also investigated the effects of Bid deficiency in cultured cells treated with anti-Fas antibody (hepatocytes and thymocytes) or with TNFalpha. (fibroblasts). In these Bid-/- cells, mitochondrial dysfunction was delayed, cytochrome c was not released, effector caspase activity was reduced and the cleavage of apoptosis substrates was altered. This loss-of-function model indicates that Bid is a critical substrate in vivo for signalling by death-receptor agonists, which mediates a mitochondrial amplification loop that is essential for the apoptosis of selected cells.
- Published
- 1999
44. BCL-2 family members and the mitochondria in apoptosis
- Author
-
Atan Gross, Stanley J. Korsmeyer, and James M. McDonnell
- Subjects
biology ,Protein Conformation ,Truncated BID ,Molecular Sequence Data ,Bcl-2 family ,Intrinsic apoptosis ,Apoptosis ,Mitochondrion ,Models, Biological ,Mitochondria ,Cell biology ,Proto-Oncogene Proteins c-bcl-2 ,Post translational ,Caspases ,Genetics ,biology.protein ,Animals ,Humans ,Amino Acid Sequence ,Apoptosome ,Protein Processing, Post-Translational ,Caspase ,Developmental Biology - Published
- 1999
45. Death and Survival Signals Determine Active/Inactive Conformations of Pro-apoptotic BAX, BAD, and BID Molecules
- Author
-
Xiao Ming Yin, Stanley J. Korsmeyer, H. Harada, Kun Wang, M. Wei, Sandra S. Zinkel, J. Zha, and Atan Gross
- Subjects
Models, Molecular ,Cell Survival ,Protein Conformation ,Biological Transport, Active ,Apoptosis ,Models, Biological ,Biochemistry ,Receptors, Tumor Necrosis Factor ,Text mining ,Antigens, CD ,Proto-Oncogene Proteins ,Genetics ,Animals ,Humans ,fas Receptor ,Phosphorylation ,Molecular Biology ,bcl-2-Associated X Protein ,Caspase 8 ,Cell Death ,Chemistry ,business.industry ,Caspase 9 ,Mitochondria ,Cell biology ,Proto-Oncogene Proteins c-bcl-2 ,Receptors, Tumor Necrosis Factor, Type I ,Caspases ,Active/Inactive ,bcl-Associated Death Protein ,Carrier Proteins ,business ,Dimerization ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Published
- 1999
46. A new Aven-ue to DNA-damage checkpoints
- Author
-
Atan Gross
- Subjects
G2 Phase ,DNA damage ,Regulator ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Xenopus laevis ,Animals ,Humans ,DNA damage checkpoints ,DNA Breaks, Double-Stranded ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Kinase ,Tumor Suppressor Proteins ,Cell Cycle ,Membrane Proteins ,Cell biology ,DNA-Binding Proteins ,Enzyme Activation ,Apoptotic Protease-Activating Factor 1 ,Apoptosis ,Apoptosis Regulatory Proteins ,BH3 Interacting Domain Death Agonist Protein - Abstract
Cells frequently arrest or die in response to DNA damage to reduce the likelihood of progression to malignancy. A recent study sheds new light on the Aven protein, a known apoptotic regulator. After DNA damage, Aven induces cell-cycle arrest via ataxia-telangiectasia-mutated (ATM) kinase activation. These findings add Aven to a growing list of apopototic regulators that function as double agents in the DNA-damage response.
- Published
- 2008
47. Regulated Targeting of BAX to Mitochondria
- Author
-
Stanley J. Korsmeyer, Ing Swie Goping, Mai Nguyen, Ronald Jemmerson, Kevin A. Roth, Gordon C. Shore, Atan Gross, and Josée N. Lavoie
- Subjects
Male ,caspase ,Poly ADP ribose polymerase ,Molecular Sequence Data ,Apoptosis ,Cysteine Proteinase Inhibitors ,Mitochondrion ,KB Cells ,Mitochondria, Heart ,Amino Acid Chloromethyl Ketones ,Rats, Sprague-Dawley ,Mice ,Cytosol ,Bcl-2-associated X protein ,Proto-Oncogene Proteins ,Animals ,Humans ,Amino Acid Sequence ,Inner mitochondrial membrane ,Cells, Cultured ,Caspase ,bcl-2-Associated X Protein ,Sequence Homology, Amino Acid ,biology ,Myocardium ,Intracellular Membranes ,Cell Biology ,Molecular biology ,Transmembrane protein ,Rats ,Cell biology ,mitochondria ,Transmembrane domain ,cytochrome c ,Proto-Oncogene Proteins c-bcl-2 ,BAX ,Caspases ,biology.protein ,Poly(ADP-ribose) Polymerases ,Sequence Alignment ,Regular Articles - Abstract
The proapoptotic protein BAX contains a single predicted transmembrane domain at its COOH terminus. In unstimulated cells, BAX is located in the cytosol and in peripheral association with intracellular membranes including mitochondria, but inserts into mitochondrial membranes after a death signal. This failure to insert into mitochondrial membrane in the absence of a death signal correlates with repression of the transmembrane signal-anchor function of BAX by the NH2-terminal domain. Targeting can be instated by deleting the domain or by replacing the BAX transmembrane segment with that of BCL-2. In stimulated cells, the contribution of the NH2 terminus of BAX correlates with further exposure of this domain after membrane insertion of the protein. The peptidyl caspase inhibitor zVAD-fmk partly blocks the stimulated mitochondrial membrane insertion of BAX in vivo, which is consistent with the ability of apoptotic cell extracts to support mitochondrial targeting of BAX in vitro, dependent on activation of caspase(s). Taken together, our results suggest that regulated targeting of BAX to mitochondria in response to a death signal is mediated by discrete domains within the BAX polypeptide. The contribution of one or more caspases may reflect an initiation and/or amplification of this regulated targeting.
- Published
- 1998
48. BID as a Double Agent in Cell Life and Death
- Author
-
Atan Gross
- Subjects
Cell cycle checkpoint ,DNA repair ,DNA damage ,Regulator ,Apoptosis ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,Animals ,Humans ,CHEK1 ,Phosphorylation ,Molecular Biology ,Tumor Suppressor Proteins ,Cell Biology ,Cell cycle ,Mitochondria ,Cell biology ,DNA-Binding Proteins ,Genes, cdc ,Cell Transformation, Neoplastic ,Tumor Suppressor Protein p53 ,BH3 Interacting Domain Death Agonist Protein ,Developmental Biology - Abstract
DNA damage leads to the activation of ATM and ATR, which in turn either cause cell cycle arrest and DNA repair or apoptosis. We have demonstrated that DNA damage leads to ATM-mediated BID phosphorylation, and that this phosphorylation regulates a novel, pro-survival function of BID important for S phase arrest. Thus, BID, a member from the core apoptotic regulatory machinery (BCL-2 family) receives direct inputs from a key regulator of the cell cycle arrest/DNA repair machinery (ATM), and therefore is an excellent candidate to coordinate genotoxic stress responses and apoptotic cell death.
- Published
- 2006
49. MTCH2: A new player in mitochondria biology
- Author
-
Atan Gross
- Subjects
Biophysics ,Cell Biology ,Mitochondrion ,Biology ,Biochemistry ,Cell biology - Published
- 2016
50. BID-D59A is a potent inducer of apoptosis in primary embryonic fibroblasts
- Author
-
Tak W. Mak, Rachel Sarig, Richard A. Flavell, Atan Gross, Richard C. Marcellus, and Yehudit Zaltsman
- Subjects
Apoptosis ,Cytochrome c Group ,Caspase 3 ,Simian virus 40 ,Bioinformatics ,Caspase 8 ,Biochemistry ,Mice ,Western blot ,medicine ,Animals ,APAF1 ,Molecular Biology ,Cells, Cultured ,Caspase-9 ,medicine.diagnostic_test ,biology ,Chemistry ,Cytochrome c ,Wild type ,Proteins ,Cell Biology ,Caspase 9 ,Mitochondria ,Cell biology ,Apoptotic Protease-Activating Factor 1 ,Caspases ,Mutation ,biology.protein ,Additions and Corrections ,Carrier Proteins ,BH3 Interacting Domain Death Agonist Protein - Abstract
The proapoptotic activity of BID seems to solely depend upon its cleavage to truncated tBID. Here we demonstrate that expression of a caspase-8 non-cleavable (nc) BID-D59A mutant or expression of wild type (wt) BID induces apoptosis in Bid -/-, caspase-8 -/-, and wt primary MEFs. Western blot analysis indicated that no cleavage products appeared in cells expressing ncBID. ncBID was as effective as wtBID in inducing cytochrome c release, caspase activation, and apoptosis. ncBID and wtBID (nc/wtBID) were much less effective than tBID in localizing to mitochondria and in inducing cytochrome c release, but only slightly less effective in inducing apoptosis. Studies with Apaf-1- and caspase-9-deficient primary MEFs indicated that both proteins were essential for nc/wtBID and for tBID-induced apoptosis. Most importantly, expression of non-apoptotic levels of either ncBID or wtBID in Bid -/- MEFs induced a similar and significant enhancement in apoptosis in response to a variety of death signals, which was accompanied by enhanced localization of BID to mitochondria and cytochrome c release. Thus, these results implicate full-length BID as an active player in the mitochondria during apoptosis.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.