Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nature ; 592(7856): 799-803, 2021 04.
Article in English | MEDLINE | ID: mdl-33854232

ABSTRACT

Mammalian development, adult tissue homeostasis and the avoidance of severe diseases including cancer require a properly orchestrated cell cycle, as well as error-free genome maintenance. The key cell-fate decision to replicate the genome is controlled by two major signalling pathways that act in parallel-the MYC pathway and the cyclin D-cyclin-dependent kinase (CDK)-retinoblastoma protein (RB) pathway1,2. Both MYC and the cyclin D-CDK-RB axis are commonly deregulated in cancer, and this is associated with increased genomic instability. The autophagic tumour-suppressor protein AMBRA1 has been linked to the control of cell proliferation, but the underlying molecular mechanisms remain poorly understood. Here we show that AMBRA1 is an upstream master regulator of the transition from G1 to S phase and thereby prevents replication stress. Using a combination of cell and molecular approaches and in vivo models, we reveal that AMBRA1 regulates the abundance of D-type cyclins by mediating their degradation. Furthermore, by controlling the transition from G1 to S phase, AMBRA1 helps to maintain genomic integrity during DNA replication, which counteracts developmental abnormalities and tumour growth. Finally, we identify the CHK1 kinase as a potential therapeutic target in AMBRA1-deficient tumours. These results advance our understanding of the control of replication-phase entry and genomic integrity, and identify the AMBRA1-cyclin D pathway as a crucial cell-cycle-regulatory mechanism that is deeply interconnected with genomic stability in embryonic development and tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin D/metabolism , Genomic Instability , S Phase , Animals , Cell Line , Cell Proliferation , Checkpoint Kinase 1/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , DNA Replication , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Humans , Mice , Mice, Knockout , Synthetic Lethal Mutations
2.
EMBO J ; 40(10): e103563, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33932238

ABSTRACT

The early secretory pathway and autophagy are two essential and evolutionarily conserved endomembrane processes that are finely interlinked. Although growing evidence suggests that intracellular trafficking is important for autophagosome biogenesis, the molecular regulatory network involved is still not fully defined. In this study, we demonstrate a crucial effect of the COPII vesicle-related protein TFG (Trk-fused gene) on ULK1 puncta number and localization during autophagy induction. This, in turn, affects formation of the isolation membrane, as well as the correct dynamics of association between LC3B and early ATG proteins, leading to the proper formation of both omegasomes and autophagosomes. Consistently, fibroblasts derived from a hereditary spastic paraparesis (HSP) patient carrying mutated TFG (R106C) show defects in both autophagy and ULK1 puncta accumulation. In addition, we demonstrate that TFG activity in autophagy depends on its interaction with the ATG8 protein LC3C through a canonical LIR motif, thereby favouring LC3C-ULK1 binding. Altogether, our results uncover a link between TFG and autophagy and identify TFG as a molecular scaffold linking the early secretion pathway to autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Proteins/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Blotting, Western , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Proteins/genetics , RNA Interference
3.
Acta Neuropathol ; 142(3): 537-564, 2021 09.
Article in English | MEDLINE | ID: mdl-34302498

ABSTRACT

Medulloblastoma (MB) is a childhood malignant brain tumour comprising four main subgroups characterized by different genetic alterations and rate of mortality. Among MB subgroups, patients with enhanced levels of the c-MYC oncogene (MBGroup3) have the poorest prognosis. Here we identify a previously unrecognized role of the pro-autophagy factor AMBRA1 in regulating MB. We demonstrate that AMBRA1 expression depends on c-MYC levels and correlates with Group 3 patient poor prognosis; also, knockdown of AMBRA1 reduces MB stem potential, growth and migration of MBGroup3 stem cells. At a molecular level, AMBRA1 mediates these effects by suppressing SOCS3, an inhibitor of STAT3 activation. Importantly, pharmacological inhibition of autophagy profoundly affects both stem and invasion potential of MBGroup3 stem cells, and a combined anti-autophagy and anti-STAT3 approach impacts the MBGroup3 outcome. Taken together, our data support the c-MYC/AMBRA1/STAT3 axis as a strong oncogenic signalling pathway with significance for both patient stratification strategies and targeted treatments of MBGroup3.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/drug effects , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Movement/genetics , Child , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells , Prognosis , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Suppressor of Cytokine Signaling 3 Protein/antagonists & inhibitors
4.
Proc Natl Acad Sci U S A ; 115(15): E3388-E3397, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581312

ABSTRACT

S-nitrosylation, a prototypic redox-based posttranslational modification, is frequently dysregulated in disease. S-nitrosoglutathione reductase (GSNOR) regulates protein S-nitrosylation by functioning as a protein denitrosylase. Deficiency of GSNOR results in tumorigenesis and disrupts cellular homeostasis broadly, including metabolic, cardiovascular, and immune function. Here, we demonstrate that GSNOR expression decreases in primary cells undergoing senescence, as well as in mice and humans during their life span. In stark contrast, exceptionally long-lived individuals maintain GSNOR levels. We also show that GSNOR deficiency promotes mitochondrial nitrosative stress, including excessive S-nitrosylation of Drp1 and Parkin, thereby impairing mitochondrial dynamics and mitophagy. Our findings implicate GSNOR in mammalian longevity, suggest a molecular link between protein S-nitrosylation and mitochondria quality control in aging, and provide a redox-based perspective on aging with direct therapeutic implications.


Subject(s)
Aging/metabolism , Mammals/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitophagy , Aging/genetics , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Cellular Senescence , Humans , Mammals/genetics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Nitric Oxide/metabolism , Nitrosative Stress , Protein Processing, Post-Translational , S-Nitrosothiols/metabolism
5.
J Neurosci ; 39(27): 5255-5268, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31043483

ABSTRACT

Lysosomal failure underlies pathogenesis of numerous congenital neurodegenerative disorders and is an early and progressive feature of Alzheimer's disease (AD) pathogenesis. Here, we report that lysosomal dysfunction in Down ayndrome (trisomy 21), a neurodevelopmental disorder and form of early onset AD, requires the extra gene copy of amyloid precursor protein (APP) and is specifically mediated by the ß cleaved carboxy terminal fragment of APP (APP-ßCTF, C99). In primary fibroblasts from individuals with DS, lysosomal degradation of autophagic and endocytic substrates is selectively impaired, causing them to accumulate in enlarged autolysosomes/lysosomes. Direct measurements of lysosomal pH uncovered a significant elevation (0.6 units) as a basis for slowed LC3 turnover and the inactivation of cathepsin D and other lysosomal hydrolases known to be unstable or less active when lysosomal pH is persistently elevated. Normalizing lysosome pH by delivering acidic nanoparticles to lysosomes ameliorated lysosomal deficits, whereas RNA sequencing analysis excluded a transcriptional contribution to hydrolase declines. Cortical neurons cultured from the Ts2 mouse model of DS exhibited lysosomal deficits similar to those in DS cells. Lowering APP expression with siRNA or BACE1 inhibition reversed cathepsin deficits in both fibroblasts and neurons. Deleting one Bace1 allele from adult Ts2 mice had similar rescue effects in vivo The modest elevation of endogenous APP-ßCTF needed to disrupt lysosomal function in DS is relevant to sporadic AD where APP-ßCTF, but not APP, is also elevated. Our results extend evidence that impaired lysosomal acidification drives progressive lysosomal failure in multiple forms of AD.SIGNIFICANCE STATEMENT Down syndrome (trisomy 21) (DS) is a neurodevelopmental disorder invariably leading to early-onset Alzheimer's disease (AD). We showed in cells from DS individuals and neurons of DS models that one extra copy of a normal amyloid precursor protein (APP) gene impairs lysosomal acidification, thereby depressing lysosomal hydrolytic activities and turnover of autophagic and endocytic substrates, processes vital to neuronal survival. These deficits, which were reversible by correcting lysosomal pH, are mediated by elevated levels of endogenous ß-cleaved carboxy-terminal fragment of APP (APP-ßCTF). Notably, similar endosomal-lysosomal pathobiology emerges early in sporadic AD, where neuronal APP-ßCTF is also elevated, underscoring its importance as a therapeutic target and underscoring the functional and pathogenic interrelationships between the endosomal-lysosomal pathway and genes causing AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Down Syndrome/metabolism , Lysosomes/metabolism , Proteolysis , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Cells, Cultured , Down Syndrome/genetics , Fibroblasts/metabolism , Humans , Peptide Fragments/metabolism
6.
Nucleic Acids Res ; 42(19): 12070-81, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25294838

ABSTRACT

Alternative splicing (AS) modulates many physiological and pathological processes. For instance, AS of the BCL-X gene balances cell survival and apoptosis in development and cancer. Herein, we identified the polypyrimidine tract binding protein (PTBP1) as a direct regulator of BCL-X AS. Overexpression of PTBP1 promotes selection of the distal 5' splice site in BCL-X exon 2, generating the pro-apoptotic BCL-Xs splice variant. Conversely, depletion of PTBP1 enhanced splicing of the anti-apoptotic BCL-XL variant. In vivo cross-linking experiments and site-directed mutagenesis restricted the PTBP1 binding site to a polypyrimidine tract located between the two alternative 5' splice sites. Binding of PTBP1 to this site was required for its effect on splicing. Notably, a similar function of PTBP1 in the selection of alternative 5' splice sites was confirmed using the USP5 gene as additional model. Mechanistically, PTBP1 displaces SRSF1 binding from the proximal 5' splice site, thus repressing its selection. Our study provides a novel mechanism of alternative 5' splice site selection by PTBP1 and indicates that the presence of a PTBP1 binding site between two alternative 5' splice sites promotes selection of the distal one, while repressing the proximal site by competing for binding of a positive regulator.


Subject(s)
Alternative Splicing , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , RNA Splice Sites , bcl-X Protein/genetics , Cell Line, Tumor , Exons , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors
7.
Oncogene ; 41(13): 1986-2002, 2022 03.
Article in English | MEDLINE | ID: mdl-35236967

ABSTRACT

Inhibitors of the mitotic kinase PLK1 yield objective responses in a subset of refractory cancers. However, PLK1 overexpression in cancer does not correlate with drug sensitivity, and the clinical development of PLK1 inhibitors has been hampered by the lack of patient selection marker. Using a high-throughput chemical screen, we discovered that cells deficient for the tumor suppressor ARID1A are highly sensitive to PLK1 inhibition. Interestingly this sensitivity was unrelated to canonical functions of PLK1 in mediating G2/M cell cycle transition. Instead, a whole-genome CRISPR screen revealed PLK1 inhibitor sensitivity in ARID1A deficient cells to be dependent on the mitochondrial translation machinery. We find that ARID1A knock-out (KO) cells have an unusual mitochondrial phenotype with aberrant biogenesis, increased oxygen consumption/expression of oxidative phosphorylation genes, but without increased ATP production. Using expansion microscopy and biochemical fractionation, we see that a subset of PLK1 localizes to the mitochondria in interphase cells. Inhibition of PLK1 in ARID1A KO cells further uncouples oxygen consumption from ATP production, with subsequent membrane depolarization and apoptosis. Knockdown of specific subunits of the mitochondrial ribosome reverses PLK1-inhibitor induced apoptosis in ARID1A deficient cells, confirming specificity of the phenotype. Together, these findings highlight a novel interphase role for PLK1 in maintaining mitochondrial fitness under metabolic stress, and a strategy for therapeutic use of PLK1 inhibitors. To translate these findings, we describe a quantitative microscopy assay for assessment of ARID1A protein loss, which could offer a novel patient selection strategy for the clinical development of PLK1 inhibitors in cancer.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Neoplasms , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Transcription Factors , Adenosine Triphosphate/metabolism , Apoptosis , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Oxygen Consumption , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Polo-Like Kinase 1
8.
Cell Death Dis ; 12(11): 1044, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728604

ABSTRACT

Autophagy is a highly dynamic and multi-step process, regulated by many functional protein units. Here, we have built up a comprehensive and up-to-date annotated gene list for the autophagy pathway, by combining previously published gene lists and the most recent publications in the field. We identified 604 genes and created main categories: MTOR and upstream pathways, autophagy core, autophagy transcription factors, mitophagy, docking and fusion, lysosome and lysosome-related genes. We then classified such genes in sub-groups, based on their functions or on their sub-cellular localization. Moreover, we have curated two shorter sub-lists to predict the extent of autophagy activation and/or lysosomal biogenesis; we next validated the "induction list" by Real-time PCR in cell lines during fasting or MTOR inhibition, identifying ATG14, ATG7, NBR1, ULK1, ULK2, and WDR45, as minimal transcriptional targets. We also demonstrated that our list of autophagy genes can be particularly useful during an effective RNA-sequencing analysis. Thus, we propose our lists as a useful toolbox for performing an informative and functionally-prognostic gene scan of autophagy steps.


Subject(s)
Autophagy/genetics , Genetic Techniques , Transcription, Genetic , Cell Line, Tumor , HEK293 Cells , Humans , Lysosomes/metabolism , Reproducibility of Results , TOR Serine-Threonine Kinases/metabolism
9.
iScience ; 24(11): 103350, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34816103

ABSTRACT

Patients with acute myeloid leukemia (AML) carrying high-risk genetic lesions or high residual disease levels after therapy are particularly exposed to the risk of relapse. Here, we identified the long non-coding RNA CDK6-AS1 able to cluster an AML subgroup with peculiar gene signatures linked to hematopoietic cell differentiation and mitochondrial dynamics. CDK6-AS1 silencing triggered hematopoietic commitment in healthy CD34+ cells, whereas in AML cells the pathological undifferentiated state was rescued. This latter phenomenon derived from RUNX1 transcriptional control, responsible for the stemness of hematopoietic precursors and for the block of differentiation in AML. By CDK6-AS1 silencing in vitro, AML mitochondrial mass decreased with augmented pharmacological sensitivity to mitochondria-targeting drugs. In vivo, the combination of tigecycline and cytarabine reduced leukemia progression in the AML-PDX model with high CDK6-AS1 levels, supporting the concept of a mitochondrial vulnerability. Together, these findings uncover CDK6-AS1 as crucial in myeloid differentiation and mitochondrial mass regulation.

10.
Cell Death Dis ; 12(3): 249, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674551

ABSTRACT

TG2 is a multifunctional enzyme involved in several cellular processes and has emerging as a potential regulator of gene expression. In this regard, we have recently shown that TG2 is able to activate HSF1, the master transcriptional regulator of the stress-responsive genes; however, its effect on the overall gene expression remains unclear. To address this point, we analyzed, by RNA-seq, the effect of TG2 on the overall transcriptome as well as we characterized the TG2 interactome in the nucleus. The data obtained from these omics approaches reveal that TG2 markedly influences the overall cellular transcriptome profile and specifically the Wnt and HSF1 pathways. In particular, its ablation leads to a drastic downregulation of many key members of these pathways. Interestingly, we found that key components of the Wnt/ß-catenin pathway are also downregulated in cells lacking HSF1, thus confirming that TG2 regulates the HSF1 and this axis controls the Wnt signaling. Mechanistic studies revealed that TG2 can regulate the Wnt pathway by physically interacts with ß-catenin and its nuclear interactome includes several proteins known to be involved in the regulation of the Wnt signaling. In order to verify whether this effect is playing a role in vivo, we ablated TG2 in Danio rerio. Our data show that the zebrafish lacking TG2 cannot complete the development and their death is associated with an evident downregulation of the Wnt pathway and a defective heat-shock response. Our findings show for the first time that TG2 is essential for the correct embryonal development of lower vertebrates, and its action is mediated by the Wnt/HSF1 axis.


Subject(s)
Fibroblasts/enzymology , GTP-Binding Proteins/metabolism , Heat Shock Transcription Factors/metabolism , Transglutaminases/metabolism , Wnt Signaling Pathway , Zebrafish/metabolism , Animals , Cells, Cultured , GTP-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Heat Shock Transcription Factors/genetics , Heat-Shock Response , Mice, Inbred C57BL , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Transcription, Genetic , Transcriptome , Transglutaminases/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
11.
Orphanet J Rare Dis ; 16(1): 112, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33648541

ABSTRACT

BACKGROUND: Adenylosuccinate lyase deficiency (ADSLD) is an ultrarare neurometabolic recessive disorder caused by loss-of-function mutations in the ADSL gene. The disease is characterized by wide clinical variability. Here we provide an updated clinical profiling of the disorder and discuss genotype-phenotype correlations. RESULTS: Data were collected through "Our Journey with ADSL deficiency Association" by using a dedicated web survey filled-in by parents. Clinical and molecular data were collected from 18 patients (12 males, median age 10.9 years ± 7.3), from 13 unrelated families. The age at onset ranged from birth to the first three years (median age 0.63 years ± 0.84 SD), and age at diagnosis varied from 2 months to 17 years, (median age 6.4 years ± 6.1 SD). The first sign was a psychomotor delay in 8/18 patients, epilepsy in 3/18, psychomotor delay and epilepsy in 3/18, and apneas, hypotonia, nystagmus in single cases. One patient (sibling of a previously diagnosed child) had a presymptomatic diagnosis. The diagnosis was made by exome sequencing in 7/18 patients. All patients were definitively diagnosed with ADSL deficiency based on pathogenic variants and/or biochemical assessment. One patient had a fatal neonatal form of ADSL deficiency, seven showed features fitting type I, and nine were characterized by a milder condition (type II), with two showing a very mild phenotype. Eighteen different variants were distributed along the entire ADSL coding sequence and were predicted to have a variable structural impact by impairing proper homotetramerization or catalytic activity of the enzyme. Six variants had not previously been reported. All but two variants were missense. CONCLUSIONS: The study adds more details on the spectrum of ADSLD patients' phenotypes and molecular data.


Subject(s)
Adenylosuccinate Lyase , Autistic Disorder , Purine-Pyrimidine Metabolism, Inborn Errors , Adenylosuccinate Lyase/deficiency , Adenylosuccinate Lyase/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Phenotype , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Purine-Pyrimidine Metabolism, Inborn Errors/genetics
12.
Nat Commun ; 12(1): 2550, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953176

ABSTRACT

Melanoma is the deadliest skin cancer. Despite improvements in the understanding of the molecular mechanisms underlying melanoma biology and in defining new curative strategies, the therapeutic needs for this disease have not yet been fulfilled. Herein, we provide evidence that the Activating Molecule in Beclin-1-Regulated Autophagy (Ambra1) contributes to melanoma development. Indeed, we show that Ambra1 deficiency confers accelerated tumor growth and decreased overall survival in Braf/Pten-mutated mouse models of melanoma. Also, we demonstrate that Ambra1 deletion promotes melanoma aggressiveness and metastasis by increasing cell motility/invasion and activating an EMT-like process. Moreover, we show that Ambra1 deficiency in melanoma impacts extracellular matrix remodeling and induces hyperactivation of the focal adhesion kinase 1 (FAK1) signaling, whose inhibition is able to reduce cell invasion and melanoma growth. Overall, our findings identify a function for AMBRA1 as tumor suppressor in melanoma, proposing FAK1 inhibition as a therapeutic strategy for AMBRA1 low-expressing melanoma.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Melanoma/genetics , Melanoma/metabolism , Animals , Autophagy/physiology , Beclin-1/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/pathology , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phenotype , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Transcriptome
13.
Mol Genet Metab Rep ; 23: 100592, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32405461

ABSTRACT

Adenylosuccinate lyase deficiency is a rare neurometabolic recessive disorder of purine metabolism characterized by a wide range of clinical manifestations. We present a very mild phenotype of two siblings characterized by mild isolated cognitive disability, in absence of brain anomalies, seizures, EEG anomalies and without progression of disease. The two patients had unsuccessfully been investigated until clinical exome was performed. In both siblings, compound heterozygosity for two inherited missense variants in ADSL gene, c.76A>T (p.Met26Leu) and c.1187G>A (p.Arg396His), were detected. Analysis of the catabolic pathway of autophagy on EBV-transformed B lymphoblastoid cell derived from the male patient excluded the presence of any autophagy alterations at the basal level. Further studies are necessary to understand the pathogenesis of the disease and to elucidate the potential role of autophagy in the development of ADSL deficiency.

14.
Cell Rep ; 32(9): 108093, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32877675

ABSTRACT

Genome editing often takes the form of either error-prone sequence disruption by non-homologous end joining (NHEJ) or sequence replacement by homology-directed repair (HDR). Although NHEJ is generally effective, HDR is often difficult in primary cells. Here, we use a combination of immunophenotyping, next-generation sequencing, and single-cell RNA sequencing to investigate and reprogram genome editing outcomes in subpopulations of adult hematopoietic stem and progenitor cells. We find that although quiescent stem-enriched cells mostly use NHEJ, non-quiescent cells with the same immunophenotype use both NHEJ and HDR. Inducing quiescence before editing results in a loss of HDR in all cell subtypes. We develop a strategy of controlled cycling and quiescence that yields a 6-fold increase in the HDR/NHEJ ratio in quiescent stem cells ex vivo and in vivo. Our results highlight the tension between editing and cellular physiology and suggest strategies to manipulate quiescent cells for research and therapeutic genome editing.


Subject(s)
CRISPR-Cas Systems/genetics , GATA3 Transcription Factor/metabolism , Gene Editing/methods , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Recombinational DNA Repair/genetics , Stem Cells/metabolism , Humans
15.
Cell Rep ; 32(5): 107993, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755585

ABSTRACT

ß-Hemoglobinopathies can trigger rapid production of red blood cells in a process known as stress erythropoiesis. Cellular stress prompts differentiating erythroid precursors to express high levels of fetal γ-globin. However, the mechanisms underlying γ-globin production during cellular stress are still poorly defined. Here, we use CRISPR-Cas genome editing to model the stress caused by reduced levels of adult ß-globin. We find that decreased ß-globin is sufficient to induce robust re-expression of γ-globin, and RNA sequencing (RNA-seq) of differentiating isogenic erythroid precursors implicates ATF4 as a causal regulator of this response. ATF4 binds within the HBS1L-MYB intergenic enhancer and regulates expression of MYB, a known γ-globin regulator. Overall, the reduction of ATF4 upon ß-globin knockout decreases the levels of MYB and BCL11A. Identification of ATF4 as a key regulator of globin compensation adds mechanistic insight to the poorly understood phenomenon of stress-induced globin compensation and could inform strategies to treat hemoglobinopathies.


Subject(s)
Activating Transcription Factor 4/metabolism , Gene Expression Regulation , Proto-Oncogene Proteins c-myb/genetics , beta-Globins/metabolism , gamma-Globins/genetics , Activating Transcription Factor 4/genetics , Base Sequence , Cell Differentiation/genetics , Cell Line , DNA, Intergenic/genetics , Down-Regulation/genetics , Enhancer Elements, Genetic/genetics , Fetal Hemoglobin/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mutation/genetics , Protein Binding , Proto-Oncogene Proteins c-myb/metabolism , Repressor Proteins/metabolism , Time Factors , Transcription, Genetic , Transcriptome/genetics , Up-Regulation/genetics , gamma-Globins/metabolism
16.
Cell Death Differ ; 26(4): 690-702, 2019 03.
Article in English | MEDLINE | ID: mdl-30728463

ABSTRACT

Autophagy and mitophagy act in cancer as bimodal processes, whose differential functions strictly depend on cancer ontogenesis, progression, and type. For instance, they can act to promote cancer progression by helping cancer cells survive stress or, instead, when mutated or abnormal, to induce carcinogenesis by influencing cell signaling or promoting intracellular toxicity. For this reason, the study of autophagy in cancer is the main focus of many researchers and several clinical trials are already ongoing to manipulate autophagy and by this way determine the outcome of disease therapy. Since the establishment of the cancer stem cell (CSC) theory and the discovery of CSCs in individual cancer types, autophagy and mitophagy have been proposed as key mechanisms in their homeostasis, dismissal or spread, even though we still miss a comprehensive view of how and by which regulatory molecules these two processes drive cell fate. In this review, we will dive into the deep water of autophagy, mitophagy, and CSCs and offer novel viewpoints on possible therapeutic strategies, based on the modulation of these degradative systems.


Subject(s)
Autophagy/drug effects , Autophagy/genetics , Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects , Animals , Autophagy/immunology , Humans , Mitophagy/drug effects , Mitophagy/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/therapy , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
Cell Death Dis ; 10(12): 868, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31740665

ABSTRACT

The endoplasmic reticulum (ER) is a key organelle fundamental for the maintenance of cellular homeostasis and to determine the cell's fate under stress conditions. Among the known proteins that regulate ER structure and function there is Reticulon-1C (RTN-1C), a member of the reticulon family localized primarily on the ER membrane. We previously demonstrated that RTN-1C expression affects ER function and stress condition. ER is an essential site for the regulation of apoptotic pathways and it has also been recently recognized as an important component of autophagic signaling. Based on these evidences, we have investigated the impact of RTN-1C modulation on autophagy induction. Interestingly we found that reticulon overexpression is able to activate autophagic machinery and its silencing results in a significative inhibition of both basal and induced autophagic response. Using different experimental approaches we demonstrated that RTN-1C colocalizes with ATG16L and LC3II on the autophagosomes. Considering the key role of reticulon proteins in the control of ER membrane shaping and homeostasis, our data suggest the participation of RTN-1C in the autophagic vesicle biogenesis at the level of the ER compartment. Our data indicate a new mechanism by which this structural ER protein modulates cellular stress, that is at the basis of different autophagy-related pathologies.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum/metabolism , Nerve Tissue Proteins/genetics , Autophagosomes/metabolism , Humans , Organelle Biogenesis
18.
Cell Death Dis ; 10(8): 563, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31332166

ABSTRACT

Down syndrome (DS), a complex genetic disorder caused by chromosome 21 trisomy, is associated with mitochondrial dysfunction leading to the accumulation of damaged mitochondria. Here we report that mitophagy, a form of selective autophagy activated to clear damaged mitochondria is deficient in primary human fibroblasts derived from individuals with DS leading to accumulation of damaged mitochondria with consequent increases in oxidative stress. We identified two molecular bases for this mitophagy deficiency: PINK1/PARKIN impairment and abnormal suppression of macroautophagy. First, strongly downregulated PARKIN and the mitophagic adaptor protein SQSTM1/p62 delays PINK1 activation to impair mitophagy induction after mitochondrial depolarization by CCCP or antimycin A plus oligomycin. Secondly, mTOR is strongly hyper-activated, which globally suppresses macroautophagy induction and the transcriptional expression of proteins critical for autophagosome formation such as ATG7, ATG3 and FOXO1. Notably, inhibition of mTOR complex 1 (mTORC1) and complex 2 (mTORC2) using AZD8055 (AZD) restores autophagy flux, PARKIN/PINK initiation of mitophagy, and the clearance of damaged mitochondria by mitophagy. These results recommend mTORC1-mTORC2 inhibition as a promising candidate therapeutic strategy for Down Syndrome.


Subject(s)
Autophagosomes/metabolism , Down Syndrome/metabolism , Mitophagy/drug effects , TOR Serine-Threonine Kinases/metabolism , Cells, Cultured , Child, Preschool , Down Syndrome/drug therapy , Down Syndrome/pathology , Fibroblasts/metabolism , Humans , Infant , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mitochondria/metabolism , Mitochondria/pathology , Morpholines/pharmacology , Morpholines/therapeutic use , Protein Kinases/metabolism , Signal Transduction/drug effects , Skin/pathology , Ubiquitin-Protein Ligases/metabolism
19.
Cell Rep ; 25(11): 3059-3073.e10, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540939

ABSTRACT

Mitochondria are key players in the regulation of T cell biology by dynamically responding to cell needs, but how these dynamics integrate in T cells is still poorly understood. We show here that the mitochondrial pro-fission protein Drp1 fosters migration and expansion of developing thymocytes both in vitro and in vivo. In addition, we find that Drp1 sustains in vitro clonal expansion and cMyc-dependent metabolic reprogramming upon activation, also regulating effector T cell numbers in vivo. Migration and extravasation defects are also exhibited in Drp1-deficient mature T cells, unveiling its crucial role in controlling both T cell recirculation in secondary lymphoid organs and accumulation at tumor sites. Moreover, the observed Drp1-dependent imbalance toward a memory-like phenotype favors T cell exhaustion in the tumor microenvironment. All of these findings support a crucial role for Drp1 in several processes during T cell development and in anti-tumor immune-surveillance.


Subject(s)
Cell Movement , Dynamins/metabolism , Immunologic Surveillance , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Count , Cell Differentiation , Cell Proliferation , Cell Survival , Homeostasis , Lymphocyte Activation/immunology , Lymphoid Tissue/metabolism , MAP Kinase Signaling System , Mice, Knockout , Phenotype , Receptors, Antigen, T-Cell , Thymocytes/cytology , Thymocytes/metabolism
20.
Front Oncol ; 7: 81, 2017.
Article in English | MEDLINE | ID: mdl-28512624

ABSTRACT

Recent decades have revealed the shape changes of mitochondria and their regulators to be main players in a plethora of physiological cell processes. Mitochondria are extremely dynamic organelles whose highly controlled morphological changes respond to specific and diverse pathophysiological needs. Thus, their qualitative control is crucial for the determination of cell function and fate. Moreover, ever-new metabolic changes, mainly attributable to mitochondrial (dys)functions, are strongly connected to cancer and its microenvironment. For this reason, the aspects controlling mitochondria activity and status are in the oncological spotlight. In this review, we elucidate the most intriguing discoveries related to two apparently independent but strictly interconnected processes crucial for the organelle functionality and fate, mitochondrial dynamics, and mitophagy. We will mostly focus on their metabolic interconnections and regulations that can causally foster a tumoral context.

SELECTION OF CITATIONS
SEARCH DETAIL