Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell ; 165(5): 1209-1223, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27133168

ABSTRACT

Across eukaryotic species, mild mitochondrial stress can have beneficial effects on the lifespan of organisms. Mitochondrial dysfunction activates an unfolded protein response (UPR(mt)), a stress signaling mechanism designed to ensure mitochondrial homeostasis. Perturbation of mitochondria during larval development in C. elegans not only delays aging but also maintains UPR(mt) signaling, suggesting an epigenetic mechanism that modulates both longevity and mitochondrial proteostasis throughout life. We identify the conserved histone lysine demethylases jmjd-1.2/PHF8 and jmjd-3.1/JMJD3 as positive regulators of lifespan in response to mitochondrial dysfunction across species. Reduction of function of the demethylases potently suppresses longevity and UPR(mt) induction, while gain of function is sufficient to extend lifespan in a UPR(mt)-dependent manner. A systems genetics approach in the BXD mouse reference population further indicates conserved roles of the mammalian orthologs in longevity and UPR(mt) signaling. These findings illustrate an evolutionary conserved epigenetic mechanism that determines the rate of aging downstream of mitochondrial perturbations.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Caenorhabditis elegans/genetics , Longevity , Mice , Mitochondria/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Unfolded Protein Response
2.
Nat Rev Mol Cell Biol ; 17(4): 213-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26956194

ABSTRACT

Mitochondria participate in crucial cellular processes such as energy harvesting and intermediate metabolism. Although mitochondria possess their own genome--a vestige of their bacterial origins and endosymbiotic evolution--most mitochondrial proteins are encoded in the nucleus. The expression of the mitochondrial proteome hence requires tight coordination between the two genomes to adapt mitochondrial function to the ever-changing cellular milieu. In this Review, we focus on the pathways that coordinate the communication between mitochondria and the nucleus during homeostasis and mitochondrial stress. These pathways include nucleus-to-mitochondria (anterograde) and mitochondria-to-nucleus (retrograde) communication, mitonuclear feedback signalling and proteostasis regulation, the integrated stress response and non-cell-autonomous communication. We discuss how mitonuclear communication safeguards cellular and organismal fitness and regulates lifespan.


Subject(s)
Cell Nucleus/physiology , Homeostasis , Mitochondria/physiology , Stress, Physiological , Animals , Cell Communication , Mitochondrial Proteins/metabolism , Models, Biological , Signal Transduction
3.
Nat Rev Mol Cell Biol ; 16(6): 345-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25970558

ABSTRACT

Recent advances in mitochondrial biology have revealed the high diversity and complexity of proteolytic enzymes that regulate mitochondrial function. We have classified mitochondrial proteases, or mitoproteases, on the basis of their function and location, and defined the human mitochondrial degradome as the complete set of mitoproteases that are encoded by the human genome. In addition to their nonspecific degradative functions, mitoproteases perform highly regulated proteolytic reactions that are important in mitochondrial function, integrity and homeostasis. These include protein synthesis, quality control, mitochondrial biogenesis and dynamics, mitophagy and apoptosis. Impaired or dysregulated function of mitoproteases is associated with ageing and with many pathological conditions such as neurodegenerative disorders, metabolic syndromes and cancer. A better understanding of the mitochondrial proteolytic landscape and its modulation may contribute to improving human lifespan and 'healthspan'.


Subject(s)
Aging/metabolism , Metabolic Syndrome/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Neurodegenerative Diseases/enzymology , Peptide Hydrolases/metabolism , Aging/genetics , Aging/pathology , Animals , Genome, Human , Humans , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Peptide Hydrolases/genetics , Proteolysis
4.
Blood ; 142(14): 1185-1192, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37506341

ABSTRACT

Germ line variants in the DDX41 gene have been linked to myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) development. However, the risks associated with different variants remain unknown, as do the basis of their leukemogenic properties, impact on steady-state hematopoiesis, and links to other cancers. Here, we investigate the frequency and significance of DDX41 variants in 454 792 United Kingdom Biobank (UKB) participants and identify 452 unique nonsynonymous DNA variants in 3538 (1/129) individuals. Many were novel, and the prevalence of most varied markedly by ancestry. Among the 1059 individuals with germ line pathogenic variants (DDX41-GPV) 34 developed MDS/AML (odds ratio, 12.3 vs noncarriers). Of these, 7 of 218 had start-lost, 22 of 584 had truncating, and 5 of 257 had missense (odds ratios: 12.9, 15.1, and 7.5, respectively). Using multivariate logistic regression, we found significant associations of DDX41-GPV with MDS, AML, and family history of leukemia but not lymphoma, myeloproliferative neoplasms, or other cancers. We also report that DDX41-GPV carriers do not have an increased prevalence of clonal hematopoiesis (CH). In fact, CH was significantly more common before sporadic vs DDX41-mutant MDS/AML, revealing distinct evolutionary paths. Furthermore, somatic mutation rates did not differ between sporadic and DDX41-mutant AML genomes, ruling out genomic instability as a driver of the latter. Finally, we found that higher mean red cell volume (MCV) and somatic DDX41 mutations in blood DNA identify DDX41-GPV carriers at increased MDS/AML risk. Collectively, our findings give new insights into the prevalence and cognate risks associated with DDX41 variants, as well as the clonal evolution and early detection of DDX41-mutant MDS/AML.


Subject(s)
GATA2 Deficiency , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Prevalence , DEAD-box RNA Helicases/genetics , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Leukemia, Myeloid, Acute/genetics , DNA
5.
EMBO J ; 39(24): e105074, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33200421

ABSTRACT

The connectivity of mitochondria is regulated by a balance between fusion and division. Many human diseases are associated with excessive mitochondrial connectivity due to impaired Drp1, a dynamin-related GTPase that mediates division. Here, we report a mitochondrial stress response, named mitochondrial safeguard, that adjusts the balance of fusion and division in response to increased mitochondrial connectivity. In cells lacking Drp1, mitochondria undergo hyperfusion. However, hyperfusion does not completely connect mitochondria because Opa1 and mitofusin 1, two other dynamin-related GTPases that mediate fusion, become proteolytically inactivated. Pharmacological and genetic experiments show that the activity of Oma1, a metalloprotease that cleaves Opa1, is regulated by short pulses of the membrane depolarization without affecting the overall membrane potential in Drp1-knockout cells. Re-activation of Opa1 and Mitofusin 1 in Drp1-knockout cells further connects mitochondria beyond hyperfusion, termed extreme fusion, leading to bioenergetic deficits. These findings reveal an unforeseen safeguard mechanism that prevents extreme fusion of mitochondria, thereby maintaining mitochondrial function when the balance is shifted to excessive connectivity.


Subject(s)
Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mitochondria/metabolism , Stress, Physiological/physiology , Animals , Dynamins/genetics , Dynamins/metabolism , Energy Metabolism , GTP Phosphohydrolases/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Metalloproteases/metabolism , Mice , Mitochondria/genetics , Mitochondrial Dynamics/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Stress, Physiological/genetics , Transcriptome
7.
Blood ; 126(2): 195-202, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26031915

ABSTRACT

Great progress has recently been achieved in the understanding of the genomic alterations driving chronic lymphocytic leukemia (CLL). Nevertheless, the specific molecular mechanisms governing chromatin remodeling in CLL are unknown. Here we report the genetic and functional characterization of somatic mutations affecting the chromatin remodeler CHD2, one of the most frequently mutated genes in CLL (5.3%) and in monoclonal B lymphocytosis (MBL, 7%), a B-cell expansion that can evolve to CLL. Most of the mutations affecting CHD2, identified by whole-exome sequencing of 456 CLL and 43 MBL patients, are either truncating or affect conserved residues in functional domains, thus supporting a putative role for CHD2 as a tumor suppressor gene. CHD2 mutants show altered nuclear distribution, and a chromodomain helicase DNA binding protein 2 (CHD2) mutant affected in its DNA-binding domain exhibits defective association with active chromatin. Clinicobiological analyses show that most CLL patients carrying CHD2 mutations also present mutated immunoglobulin heavy chain variable region genes (IGHVs), being the most frequently mutated gene in this prognostic subgroup. This is the first study providing functional evidence supporting CHD2 as a cancer driver and opens the way to further studies of the role of this chromatin remodeler in CLL.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Chromatin Assembly and Disassembly/genetics , Cohort Studies , HEK293 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Molecular Sequence Data , Sequence Homology, Amino Acid
8.
EMBO J ; 31(9): 2117-33, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22433842

ABSTRACT

Mitochondria are dynamic subcellular organelles that convert nutrient intermediates into readily available energy equivalents. Optimal mitochondrial function is ensured by a highly evolved quality control system, coordinated by protein machinery that regulates a process of continual fusion and fission. In this work, we provide in vivo evidence that the ATP-independent metalloprotease OMA1 plays an essential role in the proteolytic inactivation of the dynamin-related GTPase OPA1 (optic atrophy 1). We also show that OMA1 deficiency causes a profound perturbation of the mitochondrial fusion-fission equilibrium that has important implications for metabolic homeostasis. Thus, ablation of OMA1 in mice results in marked transcriptional changes in genes of lipid and glucose metabolic pathways and substantial alterations in circulating blood parameters. Additionally, Oma1-mutant mice exhibit an increase in body weight due to increased adipose mass, hepatic steatosis, decreased energy expenditure and impaired thermogenenesis. These alterations are especially significant under metabolic stress conditions, indicating that an intact OMA1-OPA1 system is essential for developing the appropriate adaptive response to different metabolic stressors such as a high-fat diet or cold-shock. This study provides the first description of an unexpected role in energy metabolism for the metalloprotease OMA1 and reinforces the importance of mitochondrial quality control for normal metabolic function.


Subject(s)
GTP Phosphohydrolases/metabolism , Metalloendopeptidases/deficiency , Metalloproteases/deficiency , Mitochondrial Proteins/deficiency , Obesity/metabolism , Thermogenesis/physiology , Adipocytes, Brown/metabolism , Animals , Blood Glucose/analysis , Diet, High-Fat , Embryo, Mammalian , Fibroblasts/metabolism , Lipid Metabolism , Metalloendopeptidases/genetics , Metalloproteases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/physiology , Mitochondrial Proteins/genetics
10.
Am J Physiol Renal Physiol ; 306(11): F1318-26, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24671334

ABSTRACT

Acute kidney injury (AKI) is associated with mitochondrial fragmentation, which contributes to mitochondrial damage and tubular cell apoptosis. Mitochondrial fragmentation involves the cleavage of both mitochondrial outer and inner membranes. Cleavage of the outer membrane results from Drp-1-mediated fission activation and Bak-promoted fusion arrest, but the molecular mechanism of inner membrane cleavage remains elusive. OMA1-mediated proteolysis of OPA1, a key inner membrane fusion protein, was recently suggested to account for inner membrane cleavage during cell stress. In this study, we determined the role of OMA1 in OPA1 proteolysis and mitochondrial fragmentation in experimental models of ischemic AKI. In ATP-depletion injury, knockdown of OMA1 suppressed OPA1 proteolysis, mitochondrial fragmentation, cytochrome c release, and consequent apoptosis in renal proximal tubular cells. In mice, OMA1 deficiency prevented ischemic AKI as indicated by better renal function, less tubular damage, and lower apoptosis. OPA1 proteolysis and mitochondrial injury during ischemic AKI were ameliorated in OMA1-deficient mice. Thus, OMA1-mediated OPA1 proteolysis plays an important role in the disruption of mitochondrial dynamics in ischemic AKI.


Subject(s)
Acute Kidney Injury/metabolism , GTP Phosphohydrolases/metabolism , Ischemia/metabolism , Kidney/metabolism , Metalloproteases/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Cell Line , Disease Models, Animal , GTP Phosphohydrolases/genetics , Kidney/blood supply , Male , Metalloproteases/genetics , Mice , Mitochondrial Proteins/genetics , Proteolysis , Rats
11.
Nat Genet ; 56(2): 273-280, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38233595

ABSTRACT

Myeloproliferative neoplasms (MPNs) are chronic cancers characterized by overproduction of mature blood cells. Their causative somatic mutations, for example, JAK2V617F, are common in the population, yet only a minority of carriers develop MPN. Here we show that the inherited polygenic loci that underlie common hematological traits influence JAK2V617F clonal expansion. We identify polygenic risk scores (PGSs) for monocyte count and plateletcrit as new risk factors for JAK2V617F positivity. PGSs for several hematological traits influenced the risk of different MPN subtypes, with low PGSs for two platelet traits also showing protective effects in JAK2V617F carriers, making them two to three times less likely to have essential thrombocythemia than carriers with high PGSs. We observed that extreme hematological PGSs may contribute to an MPN diagnosis in the absence of somatic driver mutations. Our study showcases how polygenic backgrounds underlying common hematological traits influence both clonal selection on somatic mutations and the subsequent phenotype of cancer.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/diagnosis , Phenotype , Janus Kinase 2/genetics , Genetic Risk Score
12.
Mol Cell Proteomics ; 10(11): M111.008094, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21828285

ABSTRACT

Lipodystrophy is a major disease involving severe alterations of adipose tissue distribution and metabolism. Mutations in genes encoding the nuclear envelope protein lamin A or its processing enzyme, the metalloproteinase Zmpste24, cause diverse human progeroid syndromes that are commonly characterized by a selective loss of adipose tissue. Similarly to humans, mice deficient in Zmpste24 accumulate prelamin A and display phenotypic features of accelerated aging, including lipodystrophy. Herein, we report the proteome and phosphoproteome of adipose tissue as well as serum metabolome in lipodystrophy by using Zmpste24(-/-) mice as experimental model. We show that Zmpste24 deficiency enhanced lipolysis, fatty acid biogenesis and ß-oxidation as well as decreased fatty acid re-esterification, thus pointing to an increased partitioning of fatty acid toward ß-oxidation and away from storage that likely underlies the observed size reduction of Zmpste24-null adipocytes. Besides the mitochondrial proteins related to lipid metabolism, other protein networks related to mitochondrial function, including those involved in tricarboxylic acid cycle and oxidative phosphorylation, were up-regulated in Zmpste24(-/-) mice. These results, together with the observation of an increased mitochondrial response to oxidative stress, support the relationship between defective prelamin A processing and mitochondrial dysfunction and highlight the relevance of oxidative damage in lipoatrophy and aging. We also show that absence of Zmpste24 profoundly alters the processing of the cytoskeletal protein vimentin and identify a novel protein dysregulated in lipodystrophy, High-Mobility Group Box-1 Protein. Finally, we found several lipid derivates with important roles in energy balance, such as Lysophosphatidylcholine or 2-arachidonoylglycerol, to be dysregulated in Zmpste24(-/-) serum. Together, our findings in Zmpste24(-/-) mice may be useful to unveil the mechanisms underlying adipose tissue dysfunction and its overall contribution to body homeostasis in progeria and other lipodystrophy syndromes as well as to develop novel strategies to prevent or ameliorate these diseases.


Subject(s)
Aging, Premature/metabolism , Lipodystrophy/metabolism , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mitochondria/metabolism , Proteome/metabolism , Vimentin/metabolism , Aging, Premature/genetics , Animals , Biomarkers/blood , Blood Glucose/metabolism , Gene Expression Regulation , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lamin Type A , Lipid Metabolism , Lipids/blood , Lipodystrophy/genetics , Male , Metabolome , Mice , Mice, Knockout , Mitochondria/enzymology , Nuclear Proteins/metabolism , Oxidation-Reduction , Phosphoproteins/metabolism , Protein Interaction Maps , Protein Precursors/metabolism , Proteome/genetics
13.
Nat Genet ; 55(9): 1523-1530, 2023 09.
Article in English | MEDLINE | ID: mdl-37620601

ABSTRACT

The myeloid neoplasms encompass acute myeloid leukemia, myelodysplastic syndromes and myeloproliferative neoplasms. Most cases arise from the shared ancestor of clonal hematopoiesis (CH). Here we analyze data from 454,340 UK Biobank participants, of whom 1,808 developed a myeloid neoplasm 0-15 years after recruitment. We describe the differences in CH mutational landscapes and hematology/biochemistry test parameters among individuals that later develop myeloid neoplasms (pre-MN) versus controls, finding that disease-specific changes are detectable years before diagnosis. By analyzing differences between 'pre-MN' and controls, we develop and validate Cox regression models quantifying the risk of progression to each myeloid neoplasm subtype. We construct 'MN-predict', a web application that generates time-dependent predictions with the input of basic blood tests and genetic data. Our study demonstrates that many individuals that develop myeloid neoplasms can be identified years in advance and provides a framework for disease-specific prognostication that will be of substantial use to researchers and physicians.


Subject(s)
Clonal Hematopoiesis , Neoplasms , Humans , Family , Mutation , Software
14.
Nat Genet ; 54(8): 1155-1166, 2022 08.
Article in English | MEDLINE | ID: mdl-35835912

ABSTRACT

Clonal hematopoiesis (CH), the clonal expansion of a blood stem cell and its progeny driven by somatic driver mutations, affects over a third of people, yet remains poorly understood. Here we analyze genetic data from 200,453 UK Biobank participants to map the landscape of inherited predisposition to CH, increasing the number of germline associations with CH in European-ancestry populations from 4 to 14. Genes at new loci implicate DNA damage repair (PARP1, ATM, CHEK2), hematopoietic stem cell migration/homing (CD164) and myeloid oncogenesis (SETBP1). Several associations were CH-subtype-specific including variants at TCL1A and CD164 that had opposite associations with DNMT3A- versus TET2-mutant CH, the two most common CH subtypes, proposing key roles for these two loci in CH development. Mendelian randomization analyses showed that smoking and longer leukocyte telomere length are causal risk factors for CH and that genetic predisposition to CH increases risks of myeloproliferative neoplasia, nonhematological malignancies, atrial fibrillation and blood epigenetic ageing.


Subject(s)
Clonal Hematopoiesis , Hematopoiesis , Cell Transformation, Neoplastic , Clonal Hematopoiesis/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Hematopoiesis/genetics , Humans , Mutation/genetics , Risk Factors
15.
J Clin Invest ; 132(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35700042

ABSTRACT

Mitochondrial stress triggers a response in the cell's mitochondria and nucleus, but how these stress responses are coordinated in vivo is poorly understood. Here, we characterize a family with myopathy caused by a dominant p.G58R mutation in the mitochondrial protein CHCHD10. To understand the disease etiology, we developed a knockin (KI) mouse model and found that mutant CHCHD10 aggregated in affected tissues, applying a toxic protein stress to the inner mitochondrial membrane. Unexpectedly, the survival of CHCHD10-KI mice depended on a protective stress response mediated by the mitochondrial metalloendopeptidase OMA1. The OMA1 stress response acted both locally within mitochondria, causing mitochondrial fragmentation, and signaled outside the mitochondria, activating the integrated stress response through cleavage of DAP3-binding cell death enhancer 1 (DELE1). We additionally identified an isoform switch in the terminal complex of the electron transport chain as a component of this response. Our results demonstrate that OMA1 was critical for neonatal survival conditionally in the setting of inner mitochondrial membrane stress, coordinating local and global stress responses to reshape the mitochondrial network and proteome.


Subject(s)
Metalloproteases , Mitochondrial Myopathies , Mitochondrial Proteins , Animals , Metalloproteases/genetics , Metalloproteases/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Myopathies/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Protein Folding
16.
Hum Mol Genet ; 17(14): 2196-211, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18443001

ABSTRACT

Autophagy is a highly regulated intracellular process involved in the turnover of most cellular constituents and in the maintenance of cellular homeostasis. It is well-established that the basal autophagic activity of living cells decreases with age, thus contributing to the accumulation of damaged macromolecules during aging. Conversely, the activity of this catabolic pathway is required for lifespan extension in animal models such as Caenorhabditis elegans and Drosophila melanogaster. In this work, we describe the unexpected finding that Zmpste24-null mice, which show accelerated aging and are a reliable model of human Hutchinson-Gilford progeria, exhibit an extensive basal activation of autophagy instead of the characteristic decline in this process occurring during normal aging. We also show that this autophagic increase is associated with a series of changes in lipid and glucose metabolic pathways, which resemble those occurring in diverse situations reported to prolong lifespan. These Zmpste24(-/-) mice metabolic alterations are also linked to substantial changes in circulating blood parameters, such as leptin, glucose, insulin or adiponectin which in turn lead to peripheral LKB1-AMPK activation and mTOR inhibition. On the basis of these results, we propose that nuclear abnormalities causing premature aging in Zmpste24(-/-) mice trigger a metabolic response involving the activation of autophagy. However, the chronic activation of this catabolic pathway may turn an originally intended pro-survival strategy into a pro-aging mechanism and could contribute to the systemic degeneration and weakening observed in these progeroid mice.


Subject(s)
Aging, Premature/physiopathology , Autophagy , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Progeria/physiopathology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Aging, Premature/genetics , Animals , Disease Models, Animal , Glucose/metabolism , Hormones/blood , Humans , Lipid Metabolism , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Progeria/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Signal Transduction
17.
Philos Trans R Soc Lond B Biol Sci ; 375(1801): 20190396, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32362267

ABSTRACT

Endosymbiotic organelles of eukaryotic cells, the plastids, including chloroplasts and mitochondria, are highly integrated into cellular signalling networks. In both heterotrophic and autotrophic organisms, plastids and/or mitochondria require extensive organelle-to-nucleus communication in order to establish a coordinated expression of their own genomes with the nuclear genome, which encodes the majority of the components of these organelles. This goal is achieved by the use of a variety of signals that inform the cell nucleus about the number and developmental status of the organelles and their reaction to changing external environments. Such signals have been identified in both photosynthetic and non-photosynthetic eukaryotes (known as retrograde signalling and retrograde response, respectively) and, therefore, appear to be universal mechanisms acting in eukaryotes of all kingdoms. In particular, chloroplasts and mitochondria both harbour crucial redox reactions that are the basis of eukaryotic life and are, therefore, especially susceptible to stress from the environment, which they signal to the rest of the cell. These signals are crucial for cell survival, lifespan and environmental adjustment, and regulate quality control and targeted degradation of dysfunctional organelles, metabolic adjustments, and developmental signalling, as well as induction of apoptosis. The functional similarities between retrograde signalling pathways in autotrophic and non-autotrophic organisms are striking, suggesting the existence of common principles in signalling mechanisms or similarities in their evolution. Here, we provide a survey for the newcomers to this field of research and discuss the importance of retrograde signalling in the context of eukaryotic evolution. Furthermore, we discuss commonalities and differences in retrograde signalling mechanisms and propose retrograde signalling as a general signalling mechanism in eukaryotic cells that will be also of interest for the specialist. This article is part of the theme issue 'Retrograde signalling from endosymbiotic organelles'.


Subject(s)
Eukaryotic Cells/metabolism , Organelles/metabolism , Signal Transduction , Symbiosis/physiology
18.
Nat Commun ; 11(1): 1407, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32179751

ABSTRACT

Leukaemogenic mutations commonly disrupt cellular differentiation and/or enhance proliferation, thus perturbing the regulatory programs that control self-renewal and differentiation of stem and progenitor cells. Translocations involving the Mll1 (Kmt2a) gene generate powerful oncogenic fusion proteins, predominantly affecting infant and paediatric AML and ALL patients. The early stages of leukaemogenic transformation are typically inaccessible from human patients and conventional mouse models. Here, we take advantage of cells conditionally blocked at the multipotent haematopoietic progenitor stage to develop a MLL-r model capturing early cellular and molecular consequences of MLL-ENL expression based on a clear clonal relationship between parental and leukaemic cells. Through a combination of scRNA-seq, ATAC-seq and genome-scale CRISPR-Cas9 screening, we identify pathways and genes likely to drive the early phases of leukaemogenesis. Finally, we demonstrate the broad utility of using matched parental and transformed cells for small molecule inhibitor studies by validating both previously known and other potential therapeutic targets.


Subject(s)
Cell Transformation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/physiopathology , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Nat Med ; 25(8): 1234-1242, 2019 08.
Article in English | MEDLINE | ID: mdl-31332389

ABSTRACT

The gut microbiome is emerging as a key regulator of several metabolic, immune and neuroendocrine pathways1,2. Gut microbiome deregulation has been implicated in major conditions such as obesity, type 2 diabetes, cardiovascular disease, non-alcoholic fatty acid liver disease and cancer3-6, but its precise role in aging remains to be elucidated. Here, we find that two different mouse models of progeria are characterized by intestinal dysbiosis with alterations that include an increase in the abundance of Proteobacteria and Cyanobacteria, and a decrease in the abundance of Verrucomicrobia. Consistent with these findings, we found that human progeria patients also display intestinal dysbiosis and that long-lived humans (that is, centenarians) exhibit a substantial increase in Verrucomicrobia and a reduction in Proteobacteria. Fecal microbiota transplantation from wild-type mice enhanced healthspan and lifespan in both progeroid mouse models, and transplantation with the verrucomicrobia Akkermansia muciniphila was sufficient to exert beneficial effects. Moreover, metabolomic analysis of ileal content points to the restoration of secondary bile acids as a possible mechanism for the beneficial effects of reestablishing a healthy microbiome. Our results demonstrate that correction of the accelerated aging-associated intestinal dysbiosis is beneficial, suggesting the existence of a link between aging and the gut microbiota that provides a rationale for microbiome-based interventions against age-related diseases.


Subject(s)
Fecal Microbiota Transplantation , Longevity , Progeria/therapy , Animals , Disease Models, Animal , Dysbiosis , Female , Gastrointestinal Microbiome , Humans , Male , Metabolomics , Mice , Mice, Inbred C57BL
20.
Methods Mol Biol ; 1731: 49-56, 2018.
Article in English | MEDLINE | ID: mdl-29318542

ABSTRACT

Mitochondrial aconitase is a reversible enzyme that catalyzes the conversion of citrate to isocitrate in the tricarboxylic acid cycle. Mitochondrial aconitase is very sensitive to oxidative inactivation and can aggregate and accumulate in the mitochondrial matrix causing mitochondrial dysfunction. Lon protease, one of the major quality control proteases in mitochondria, degrades oxidized aconitase maintaining mitochondrial homeostasis. This chapter describes a step-by-step protocol for a simple and reliable measurement of mitochondrial aconitase, as well as citrate synthase activity, using isolated mitochondria from cells. The protocol is simple and fast, and it is optimized for a 96-well plate using a microplate reader.


Subject(s)
ATP-Dependent Proteases/metabolism , Aconitate Hydratase/analysis , Enzyme Assays/methods , Mitochondrial Proteins/metabolism , Aconitate Hydratase/metabolism , Animals , Cell Line, Tumor , Citrate (si)-Synthase/analysis , Citrate (si)-Synthase/metabolism , Enzyme Assays/instrumentation , Fibroblasts , Mice , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Primary Cell Culture
SELECTION OF CITATIONS
SEARCH DETAIL