Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 50
Filtrer
1.
Cells ; 13(10)2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38786071

RÉSUMÉ

The risk of aberrant growth of induced pluripotent stem cell (iPSC)-derived cells in response to DNA damage is a potential concern as the tumor suppressor genes TP53 and CDKN2A are transiently inactivated during reprogramming. Herein, we evaluate the integrity of cellular senescence pathways and DNA double-strand break (DSB) repair in Sendai virus reprogrammed iPSC-derived human fibroblasts (i-HF) compared to their parental skin fibroblasts (HF). Using transcriptomics analysis and a variety of functional assays, we show that the capacity of i-HF to enter senescence and repair DSB is not compromised after damage induced by ionizing radiation (IR) or the overexpression of H-RASV12. Still, i-HF lines are transcriptionally different from their parental lines, showing enhanced metabolic activity and higher expression of p53-related effector genes. As a result, i-HF lines generally exhibit increased sensitivity to various stresses, have an elevated senescence-associated secretory phenotype (SASP), and cannot be immortalized unless p53 expression is knocked down. In conclusion, while our results suggest that i-HF are not at a greater risk of transformation, their overall hyperactivation of senescence pathways may impede their function as a cell therapy product.


Sujet(s)
Vieillissement de la cellule , Fibroblastes , Cellules souches pluripotentes induites , Humains , Fibroblastes/métabolisme , Cellules souches pluripotentes induites/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Réparation de l'ADN , Cassures double-brin de l'ADN , Stress physiologique , Reprogrammation cellulaire , Rayonnement ionisant
2.
Nat Commun ; 15(1): 2435, 2024 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-38499573

RÉSUMÉ

The potential of immune checkpoint inhibitors (ICI) may be limited in situations where immune cell fitness is impaired. Here, we show that the efficacy of cancer immunotherapies is compromised by the accumulation of senescent cells in mice and in the context of therapy-induced senescence (TIS). Resistance to immunotherapy is associated with a decrease in the accumulation and activation of CD8 T cells within tumors. Elimination of senescent cells restores immune homeostasis within the tumor micro-environment (TME) and increases mice survival in response to immunotherapy. Using single-cell transcriptomic analysis, we observe that the injection of ABT263 (Navitoclax) reverses the exacerbated immunosuppressive profile of myeloid cells in the TME. Elimination of these myeloid cells also restores CD8 T cell proliferation in vitro and abrogates immunotherapy resistance in vivo. Overall, our study suggests that the use of senolytic drugs before ICI may constitute a pharmacological approach to improve the effectiveness of cancer immunotherapies.


Sujet(s)
Tumeurs , Microenvironnement tumoral , Animaux , Souris , Immunothérapie , Tumeurs/anatomopathologie , Vieillissement de la cellule
3.
Nat Commun ; 14(1): 4033, 2023 07 19.
Article de Anglais | MEDLINE | ID: mdl-37468473

RÉSUMÉ

Muscle stem cells, the engine of muscle repair, are affected in myotonic dystrophy type 1 (DM1); however, the underlying molecular mechanism and the impact on the disease severity are still elusive. Here, we show using patients' samples that muscle stem cells/myoblasts exhibit signs of cellular senescence in vitro and in situ. Single cell RNAseq uncovers a subset of senescent myoblasts expressing high levels of genes related to the senescence-associated secretory phenotype (SASP). We show that the levels of interleukin-6, a prominent SASP cytokine, in the serum of DM1 patients correlate with muscle weakness and functional capacity limitations. Drug screening revealed that the senolytic BCL-XL inhibitor (A1155463) can specifically remove senescent DM1 myoblasts by inducing their apoptosis. Clearance of senescent cells reduced the expression of SASP, which rescued the proliferation and differentiation capacity of DM1 myoblasts in vitro and enhanced their engraftment following transplantation in vivo. Altogether, this study identifies the pathogenic mechanism associated with muscle stem cell defects in DM1 and opens a therapeutic avenue that targets these defective cells to restore myogenesis.


Sujet(s)
Dystrophie myotonique , Cellules satellites du muscle squelettique , Humains , Dystrophie myotonique/traitement médicamenteux , Dystrophie myotonique/génétique , Dystrophie myotonique/métabolisme , Sénothérapie , Fibres musculaires squelettiques/métabolisme , Cellules satellites du muscle squelettique/métabolisme , Développement musculaire/génétique
4.
Cell Rep ; 40(7): 111241, 2022 08 16.
Article de Anglais | MEDLINE | ID: mdl-35977509

RÉSUMÉ

Previous reports showed that mouse vaccination with pluripotent stem cells (PSCs) induces durable anti-tumor immune responses via T cell recognition of some elusive oncofetal epitopes. We characterize the MHC I-associated peptide (MAP) repertoire of human induced PSCs (iPSCs) using proteogenomics. Our analyses reveal a set of 46 pluripotency-associated MAPs (paMAPs) absent from the transcriptome of normal tissues and adult stem cells but expressed in PSCs and multiple adult cancers. These paMAPs derive from coding and allegedly non-coding (48%) transcripts involved in pluripotency maintenance, and their expression in The Cancer Genome Atlas samples correlates with source gene hypomethylation and genomic aberrations common across cancer types. We find that several of these paMAPs were immunogenic. However, paMAP expression in tumors coincides with activation of pathways instrumental in immune evasion (WNT, TGF-ß, and CDK4/6). We propose that currently available inhibitors of these pathways could synergize with immune targeting of paMAPs for the treatment of poorly differentiated cancers.


Sujet(s)
Cellules souches pluripotentes induites , Tumeurs , Cellules souches pluripotentes , Animaux , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Souris , Tumeurs/métabolisme , Peptides/métabolisme , Cellules souches pluripotentes/métabolisme
5.
Cell Rep Methods ; 2(1): 100153, 2022 01 24.
Article de Anglais | MEDLINE | ID: mdl-35474871

RÉSUMÉ

Modeling the tumor-immune cell interactions in humanized mice is complex and limits drug development. Here, we generated easily accessible tumor models by transforming either primary skin fibroblasts or induced pluripotent stem cell-derived cell lines injected in immune-deficient mice reconstituted with human autologous immune cells. Our results showed that fibroblastic, hepatic, or neural tumors were all efficiently infiltrated and partially or totally rejected by autologous immune cells in humanized mice. Characterization of tumor-immune infiltrates revealed high expression levels of the dysfunction markers Tim3 and PD-1 in T cells and an enrichment in regulatory T cells, suggesting rapid establishment of immunomodulatory phenotypes. Inhibition of PD-1 by Nivolumab in humanized mice resulted in increased immune cell infiltration and a slight decrease in tumor growth. We expect that these versatile and accessible cancer models will facilitate preclinical studies and the evaluation of autologous cancer immunotherapies across a range of different tumor cell types.


Sujet(s)
Cellules souches pluripotentes induites , Tumeurs , Souris , Humains , Animaux , Cellules souches pluripotentes induites/métabolisme , Récepteur-1 de mort cellulaire programmée , Tumeurs/thérapie , Nivolumab , Immunothérapie/méthodes
6.
Pharmacogenomics ; 23(7): 415-430, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35485735

RÉSUMÉ

Aims: To investigate the role of MYBBP1A gene and rs3809849 in pancreatic cancer (PANC1) and lymphoblastic leukemia (NALM6) cell lines and their response to asparaginase treatment. Materials & methods: The authors applied CRISPR-Cas9 to produce MYBBP1A knock-out (KO) and rs3809849 knock-in (KI) cell lines. The authors also interrogated rs3809849's impact on PANC1 cells through allele-specific overexpression. Results: PANC1 MYBBP1A KO cells exhibited lower proliferation capacity (p ≤ 0.05), higher asparaginase sensitivity (p = 0.01), reduced colony-forming potential (p = 0.001), cell cycle blockage in S phase, induction of apoptosis and remarkable morphology changes suggestive of an epithelial-mesenchymal transition. Overexpression of the wild-type (but not the mutant) allele of MYBBP1A-rs3809849 in PANC1 cells increased asparaginase sensitivity. NALM6 MYBBP1A KO displayed resistance to asparaginase (p < 0.0001), whereas no effect for rs3809849 KI was noted. Conclusions:MYBBP1A is important for regulating various cellular functions, and it plays, along with its rs3809849 polymorphism, a tissue-specific role in asparaginase treatment response.


Sujet(s)
Tumeurs du pancréas , Leucémie-lymphome lymphoblastique à précurseurs B et T , Allèles , Asparaginase/génétique , Asparaginase/pharmacologie , Asparaginase/usage thérapeutique , Protéines de liaison à l'ADN/génétique , Humains , Tumeurs du pancréas/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Protéines de liaison à l'ARN/génétique , Facteurs de transcription/génétique , Tumeurs du pancréas
7.
Cells ; 10(7)2021 06 22.
Article de Anglais | MEDLINE | ID: mdl-34206425

RÉSUMÉ

Cancer incidence increases drastically with age. Of the many possible reasons for this, there is the accumulation of senescent cells in tissues and the loss of function and proliferation potential of immune cells, often referred to as immuno-senescence. Immune checkpoint inhibitors (ICI), by invigorating immune cells, have the potential to be a game-changers in the treatment of cancer. Yet, the variability in the efficacy of ICI across patients and cancer types suggests that several factors influence the success of such inhibitors. There is currently a lack of clinical studies measuring the impact of aging and senescence on ICI-based therapies. Here, we review how cellular senescence and aging, either by directly altering the immune system fitness or indirectly through the modification of the tumor environment, may influence the cancer-immune response.


Sujet(s)
Vieillissement/anatomopathologie , Vieillissement de la cellule , Immunothérapie , Tumeurs/immunologie , Tumeurs/thérapie , Humains , Modèles biologiques , Résultat thérapeutique
8.
Radiat Res ; 196(3): 315-322, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-34107047

RÉSUMÉ

Decreased neurogenesis after brain exposure to ionizing radiation is linked to neurocognitive impairments. Using transgenic mouse models, we previously showed that abrogation of radiation-induced senescence, or apoptosis, can partially rescue neurogenesis in the subventricular and hippocampus regions. Here, we evaluate whether the injection of recombinant epidermal growth factor (rEGF) or mesenchymal stromal cells (MSC) engineered to secrete EGF (MSC-EGF) can preserve neurogenesis. Using doublecortin (Dcx) expression and BrdU incorporation assays, we found that the injection of rEGF into the subventricular zone (SVZ) promotes neurogenesis, despite increasing apoptosis, in the brain of irradiated mice. The effect of rEGF was mostly localized, as Dcx expression was not induced in the hippocampus region and limited in the contralateral SVZ. Surprisingly, the injection of bone marrow-derived MSC alone, or secreting EGF, did not result in increased neurogenesis despite the fact that part of the MSC survived a few weeks after injection. Our results suggest that only a supraphysiological concentration of rEGF can promote neurogenesis, likely through a direct mitogenic effect.


Sujet(s)
Facteur de croissance épidermique/usage thérapeutique , Transplantation de cellules souches mésenchymateuses/méthodes , Neurogenèse , Radioprotecteurs/usage thérapeutique , Animaux , Irradiation crânienne/effets indésirables , Protéine doublecortine , Facteur de croissance épidermique/administration et posologie , Facteur de croissance épidermique/génétique , Femelle , Vecteurs génétiques/génétique , Injections ventriculaires , Lentivirus/génétique , Cellules souches mésenchymateuses/métabolisme , Souris , Souris de lignée C57BL , Neurogenèse/effets des médicaments et des substances chimiques , Radioprotecteurs/administration et posologie , Protéines recombinantes/administration et posologie , Protéines recombinantes/usage thérapeutique , Transduction génétique
9.
Cell Metab ; 33(4): 818-832.e7, 2021 04 06.
Article de Anglais | MEDLINE | ID: mdl-33548171

RÉSUMÉ

Attenuating pathological angiogenesis in diseases characterized by neovascularization such as diabetic retinopathy has transformed standards of care. Yet little is known about the molecular signatures discriminating physiological blood vessels from their diseased counterparts, leading to off-target effects of therapy. We demonstrate that in contrast to healthy blood vessels, pathological vessels engage pathways of cellular senescence. Senescent (p16INK4A-expressing) cells accumulate in retinas of patients with diabetic retinopathy and during peak destructive neovascularization in a mouse model of retinopathy. Using either genetic approaches that clear p16INK4A-expressing cells or small molecule inhibitors of the anti-apoptotic protein BCL-xL, we show that senolysis suppresses pathological angiogenesis. Single-cell analysis revealed that subsets of endothelial cells with senescence signatures and expressing Col1a1 are no longer detected in BCL-xL-inhibitor-treated retinas, yielding a retina conducive to physiological vascular repair. These findings provide mechanistic evidence supporting the development of BCL-xL inhibitors as potential treatments for neovascular retinal disease.


Sujet(s)
Vieillissement de la cellule , Rétinopathies/anatomopathologie , Protéine bcl-X/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Chaine alpha-1 du collagène de type I/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/déficit , Inhibiteur p16 de kinase cycline-dépendante/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Modèles animaux de maladie humaine , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Femelle , Flavonols/composition chimique , Flavonols/pharmacologie , Flavonols/usage thérapeutique , Humains , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Néovascularisation pathologique , Rétinopathies/traitement médicamenteux , Rétinopathies/métabolisme , Tacrolimus/analogues et dérivés , Tacrolimus/pharmacologie , Protéine bcl-X/antagonistes et inhibiteurs
10.
Stem Cells Transl Med ; 10(2): 267-277, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32881406

RÉSUMÉ

It is still unclear if immune responses will compromise the large-scale utilization of human induced pluripotent stem cells (hiPSCs)-derived cell therapies. To answer this question, we used humanized mouse models generated by the adoptive transfer of peripheral blood mononuclear cells or the cotransplantation of hematopoietic stem cells and human thymic tissue. Using these mice, we evaluated the engraftment in skeletal muscle of myoblasts derived either directly from a muscle biopsy or differentiated from hiPSCs or fibroblasts. Our results showed that while allogeneic grafts were mostly rejected and highly infiltrated with human T cells, engraftment of autologous cells was tolerated. We also observed that hiPSC-derived myogenic progenitor cells (MPCs) are not targeted by autologous T cells and natural killer cells in vitro. These findings suggest that the reprogramming and differentiation procedures we used are not immunogenic and that hiPSC-derived MPCs will be tolerated in the presence of a competent human immune system.


Sujet(s)
Cellules souches pluripotentes induites , Transfert adoptif , Animaux , Différenciation cellulaire , Reprogrammation cellulaire , Fibroblastes , Transplantation de cellules souches hématopoïétiques , Humains , Cellules souches pluripotentes induites/transplantation , Agranulocytes , Souris , Myoblastes , Thymus (glande)/cytologie
11.
Nat Commun ; 11(1): 4979, 2020 10 05.
Article de Anglais | MEDLINE | ID: mdl-33020468

RÉSUMÉ

Cellular senescence is a known driver of carcinogenesis and age-related diseases, yet senescence is required for various physiological processes. However, the mechanisms and factors that control the negative effects of senescence while retaining its benefits are still elusive. Here, we show that the rasGAP SH3-binding protein 1 (G3BP1) is required for the activation of the senescent-associated secretory phenotype (SASP). During senescence, G3BP1 achieves this effect by promoting the association of the cyclic GMP-AMP synthase (cGAS) with cytosolic chromatin fragments. In turn, G3BP1, through cGAS, activates the NF-κB and STAT3 pathways, promoting SASP expression and secretion. G3BP1 depletion or pharmacological inhibition impairs the cGAS-pathway preventing the expression of SASP factors without affecting cell commitment to senescence. These SASPless senescent cells impair senescence-mediated growth of cancer cells in vitro and tumor growth in vivo. Our data reveal that G3BP1 is required for SASP expression and that SASP secretion is a primary mediator of senescence-associated tumor growth.


Sujet(s)
Vieillissement de la cellule/physiologie , Helicase/métabolisme , Tumeurs/anatomopathologie , Protéines liant le poly-adp-ribose/métabolisme , RNA helicases/métabolisme , Protéines à motif de reconnaissance de l'ARN/métabolisme , Cellules A549 , Animaux , Carcinogenèse , Lignée cellulaire , Mouvement cellulaire , Cytokines/métabolisme , Helicase/antagonistes et inhibiteurs , Helicase/déficit , Humains , Inflammation , Souris , Tumeurs/métabolisme , Nucleotidyltransferases/métabolisme , Protéines liant le poly-adp-ribose/antagonistes et inhibiteurs , Protéines liant le poly-adp-ribose/déficit , RNA helicases/antagonistes et inhibiteurs , RNA helicases/déficit , Protéines à motif de reconnaissance de l'ARN/antagonistes et inhibiteurs , Protéines à motif de reconnaissance de l'ARN/déficit , Facteur de transcription STAT-3/métabolisme , Transduction du signal , Facteur de transcription RelA/métabolisme
12.
Front Immunol ; 10: 2580, 2019.
Article de Anglais | MEDLINE | ID: mdl-31787975

RÉSUMÉ

The safe utilization of induced pluripotent stem cell (iPSC) derivatives in clinical use is attributed to the complete elimination of the risk of forming teratomas after transplantation. The extent by which such a risk exists in immune-competent hosts is mostly unknown. Here, using humanized mice reconstituted with fetal hematopoietic stem cells and autologous thymus tissue (bone-liver-thymus humanized mice [Hu-BLT]) or following the adoptive transfer of peripheral blood mononuclear cells(PBMCs) (Hu-AT), we evaluated the capacity of immune cells to prevent or eliminate teratomas derived from human iPSCs (hiPSCs). Our results showed that the injection of hiPSCs failed to form teratomas in Hu-AT mice reconstituted with allogeneic or autologous PBMCs or purified natural killer (NK) cells alone. However, teratomas were observed in Hu-AT mice reconstituted with autologous PBMCs depleted from NK cells. In line with these results, Hu-BLT, which do not have functional NK cells, could not prevent the growth of teratomas. Finally, we found that established teratomas were not targeted by NK cells and instead were efficiently rejected by allogeneic but not autologous T cells in Hu-AT mice. Overall, our findings suggest that autologous hiPSC-derived therapies are unlikely to form teratomas in the presence of NK cells.


Sujet(s)
Cellules tueuses naturelles/immunologie , Cellules souches pluripotentes/immunologie , Tératome/prévention et contrôle , Transfert adoptif/effets indésirables , Adulte , Animaux , Humains , Agranulocytes/transplantation , Souris , Souris de lignée NOD , Souris SCID , Lymphocytes T/immunologie , Tératome/étiologie , Tératome/immunologie , Transplantation hétérologue
13.
J Immunol ; 203(10): 2735-2745, 2019 11 15.
Article de Anglais | MEDLINE | ID: mdl-31578272

RÉSUMÉ

Therapeutic uses of mesenchymal stromal cells (MSCs) have emerged over the past decade. Yet, their effect on tumor growth remains highly debated, particularly in an immune competent environment. In this study, we wanted to investigate the impact of human umbilical cord-derived MSCs (hUC-MSCs) on tumor growth in humanized mice generated by the human adoptive transfer of PBMCs or the cotransplantation of hematopoietic stem cells and human thymic tissue (human BLT [Hu-BLT]). Our results showed that the growth and immune rejection of engineered human fibroblastic tumors was not altered by the injection of hUC-MSCs in immune-deficient or humanized mice, respectively. This was observed whether tumor cells were injected s.c. or i.v. and independently of the injection route of the hUC-MSCs. Moreover, only in Hu-BLT mice did hUC-MSCs have some effects on the tumor-immune infiltrate, yet without altering tumor growth. These results demonstrate that hUC-MSCs do not promote fibroblastic tumor growth and neither do they prevent tumor infiltration and rejection by immune cells in humanized mice.


Sujet(s)
Lymphocytes TIL/immunologie , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/immunologie , Transfert adoptif , Animaux , Lignée de cellules transformées/transplantation , Fibroblastes/transplantation , Vecteurs génétiques , Rejet du greffon/immunologie , Hétérogreffes , Humains , Injections veineuses , Injections sous-cutanées , Souris , Souris de lignée NOD , Souris SCID , Transplantation tumorale , Tumeurs expérimentales/immunologie , Chimère post-radique , Organismes exempts d'organismes pathogènes spécifiques , Telomerase/génétique , Telomerase/physiologie , Thymus (glande)/transplantation , Gelée de Wharton/cytologie
15.
J Exp Clin Cancer Res ; 38(1): 251, 2019 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-31196146

RÉSUMÉ

BACKGROUND: Cardiac glycosides are approved for the treatment of heart failure as Na+/K+ pump inhibitors. Their repurposing in oncology is currently investigated in preclinical and clinical studies. However, the identification of a specific cancer type defined by a molecular signature to design targeted clinical trials with cardiac glycosides remains to be characterized. Here, we demonstrate that cardiac glycoside proscillaridin A specifically targets MYC overexpressing leukemia cells and leukemia stem cells by causing MYC degradation, epigenetic reprogramming and leukemia differentiation through loss of lysine acetylation. METHODS: Proscillaridin A anticancer activity was investigated against a panel of human leukemia and solid tumor cell lines with different MYC expression levels, overexpression in vitro systems and leukemia stem cells. RNA-sequencing and differentiation studies were used to characterize transcriptional and phenotypic changes. Drug-induced epigenetic changes were studied by chromatin post-translational modification analysis, expression of chromatin regulators, chromatin immunoprecipitation, and mass-spectrometry. RESULTS: At a clinically relevant dose, proscillaridin A rapidly altered MYC protein half-life causing MYC degradation and growth inhibition. Transcriptomic profile of leukemic cells after treatment showed a downregulation of genes involved in MYC pathways, cell replication and an upregulation of hematopoietic differentiation genes. Functional studies confirmed cell cycle inhibition and the onset of leukemia differentiation even after drug removal. Proscillaridin A induced a significant loss of lysine acetylation in histone H3 (at lysine 9, 14, 18 and 27) and in non-histone proteins such as MYC itself, MYC target proteins, and a series of histone acetylation regulators. Global loss of acetylation correlated with the rapid downregulation of histone acetyltransferases. Importantly, proscillaridin A demonstrated anticancer activity against lymphoid and myeloid stem cell populations characterized by MYC overexpression. CONCLUSION: Overall, these results strongly support the repurposing of proscillaridin A in MYC overexpressing leukemia.


Sujet(s)
Antinéoplasiques/effets indésirables , Expression des gènes/effets des médicaments et des substances chimiques , Gènes myc , Défaillance cardiaque/étiologie , Leucémies/génétique , Lysine/métabolisme , Proscillaridine/effets indésirables , Acétylation , Antinéoplasiques/usage thérapeutique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chromatine/génétique , Chromatine/métabolisme , Relation dose-effet des médicaments , Épigenèse génétique/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes , Histone/métabolisme , Humains , Leucémies/complications , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Modèles biologiques , Proscillaridine/usage thérapeutique , Lymphocytes T/cytologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme
16.
Aging Cell ; 18(4): e12971, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31148373

RÉSUMÉ

Some studies show eliminating senescent cells rejuvenate aged mice and attenuate deleterious effects of chemotherapy. Nevertheless, it remains unclear whether senescence affects immune cell function. We provide evidence that exposure of mice to ionizing radiation (IR) promotes the senescent-associated secretory phenotype (SASP) and expression of p16INK4a in splenic cell populations. We observe splenic T cells exhibit a reduced proliferative response when cultured with allogenic cells in vitro and following viral infection in vivo. Using p16-3MR mice that allow elimination of p16INK4a -positive cells with exposure to ganciclovir, we show that impaired T-cell proliferation is partially reversed, mechanistically dependent on p16INK4a expression and the SASP. Moreover, we found macrophages isolated from irradiated spleens to have a reduced phagocytosis activity in vitro, a defect also restored by the elimination of p16INK4a expression. Our results provide molecular insight on how senescence-inducing IR promotes loss of immune cell fitness, which suggest senolytic drugs may improve immune cell function in aged and patients undergoing cancer treatment.


Sujet(s)
Vieillissement de la cellule/effets des radiations , Rayonnement ionisant , Rate/métabolisme , Rate/effets des radiations , Lymphocytes T/immunologie , Lymphocytes T/effets des radiations , Animaux , Antiviraux/usage thérapeutique , Infections à Arenaviridae/traitement médicamenteux , Infections à Arenaviridae/immunologie , Infections à Arenaviridae/virologie , Prolifération cellulaire/effets des radiations , Cellules cultivées , Vieillissement de la cellule/génétique , Inhibiteur p16 de kinase cycline-dépendante/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Ganciclovir/usage thérapeutique , Virus de la chorioméningite lymphocytaire/immunologie , Macrophages/métabolisme , Souris , Souris transgéniques , Phénotype , Rajeunissement/physiologie , Rate/virologie
17.
Nature ; 566(7742): 73-78, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30728521

RÉSUMÉ

Retrotransposable elements are deleterious at many levels, and the failure of host surveillance systems for these elements can thus have negative consequences. However, the contribution of retrotransposon activity to ageing and age-associated diseases is not known. Here we show that during cellular senescence, L1 (also known as LINE-1) retrotransposable elements become transcriptionally derepressed and activate a type-I interferon (IFN-I) response. The IFN-I response is a phenotype of late senescence and contributes to the maintenance of the senescence-associated secretory phenotype. The IFN-I response is triggered by cytoplasmic L1 cDNA, and is antagonized by inhibitors of the L1 reverse transcriptase. Treatment of aged mice with the nucleoside reverse transcriptase inhibitor lamivudine downregulated IFN-I activation and age-associated inflammation (inflammaging) in several tissues. We propose that the activation of retrotransposons is an important component of sterile inflammation that is a hallmark of ageing, and that L1 reverse transcriptase is a relevant target for the treatment of age-associated disorders.


Sujet(s)
Vieillissement de la cellule/génétique , Inflammation/génétique , Interféron de type I/métabolisme , Éléments LINE/génétique , Vieillissement/génétique , Vieillissement/anatomopathologie , Animaux , Régulation négative , Femelle , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Humains , Inflammation/anatomopathologie , Lamivudine/pharmacologie , Mâle , Souris , Phénotype , RNA-directed DNA polymerase/génétique , RNA-directed DNA polymerase/métabolisme , Inhibiteurs de la transcriptase inverse/pharmacologie
18.
Stem Cell Reports ; 10(6): 1721-1733, 2018 06 05.
Article de Anglais | MEDLINE | ID: mdl-29706499

RÉSUMÉ

Brain neurogenesis is severely impaired following exposure to ionizing radiation (IR). We and others have shown that the expression of the tumor suppressor gene p16INK4a is increased in tissues exposed to IR and thus hypothesized that its expression could limit neurogenesis in the irradiated brain. Here, we found that exposure to IR leads to persistent DNA damage and the expression of p16INK4a in the hippocampus and subventricular zone regions. This was accompanied by a decline in neurogenesis, as determined by doublecortin expression and bromodeoxyuridine incorporation, an effect partially restored in Ink4a/arf-null mice. Increased neurogenesis in the absence of INK4a/ARF expression was independent of apoptosis and activation of the microglia. Moreover, treatment of irradiated mice with a superoxide dismutase mimetic or clearance of p16INK4a-expressing cells using mouse genetics failed to increase neurogenesis. In conclusion, our results suggest that IR-induced p16INK4a expression is a mechanism that limits neurogenesis.


Sujet(s)
Encéphale/métabolisme , Encéphale/effets des radiations , Inhibiteur p16 de kinase cycline-dépendante/génétique , Régulation de l'expression des gènes/effets des radiations , Neurogenèse/génétique , Neurogenèse/effets des radiations , Rayonnement ionisant , Animaux , Apoptose/génétique , Marqueurs biologiques , Biomimétique , Encéphale/anatomopathologie , Vieillissement de la cellule/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Altération de l'ADN/effets des radiations , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Souris , Souris transgéniques , Microglie/métabolisme , Imagerie moléculaire , Cellules souches neurales/cytologie , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Cellules souches neurales/effets des radiations , Neurogenèse/effets des médicaments et des substances chimiques , Dose de rayonnement , Superoxide dismutase/métabolisme , Superoxide dismutase/pharmacologie , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
19.
Aging Cell ; 16(2): 422-427, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28124509

RÉSUMÉ

The DNA damage response (DDR) arrests cell cycle progression until DNA lesions, like DNA double-strand breaks (DSBs), are repaired. The presence of DSBs in cells is usually detected by indirect techniques that rely on the accumulation of proteins at DSBs, as part of the DDR. Such detection may be biased, as some factors and their modifications may not reflect physical DNA damage. The dependency on DDR markers of DSB detection tools has left questions unanswered. In particular, it is known that senescent cells display persistent DDR foci, that we and others have proposed to be persistent DSBs, resistant to endogenous DNA repair activities. Others have proposed that these peculiar DDR foci might not be sites of damaged DNA per se but instead stable chromatin modifications, termed DNA-SCARS. Here, we developed a method, named 'DNA damage in situ ligation followed by proximity ligation assay' (DI-PLA) for the detection and imaging of DSBs in cells. DI-PLA is based on the capture of free DNA ends in fixed cells in situ, by ligation to biotinylated double-stranded DNA oligonucleotides, which are next recognized by antibiotin anti-bodies. Detection is enhanced by PLA with a partner DDR marker at the DSB. We validated DI-PLA by demonstrating its ability to detect DSBs induced by various genotoxic insults in cultured cells and tissues. Most importantly, by DI-PLA, we demonstrated that both senescent cells in culture and tissues from aged mammals retain true unrepaired DSBs associated with DDR markers.


Sujet(s)
Vieillissement/anatomopathologie , Vieillissement de la cellule , Altération de l'ADN , Mammifères/physiologie , Analyse sur cellule unique/méthodes , Animaux , Lignée cellulaire tumorale , Vieillissement de la cellule/effets des radiations , Cassures double-brin de l'ADN/effets des radiations , Humains , Souris , Réaction de polymérisation en chaîne , Rayonnement ionisant
20.
Oncotarget ; 7(21): 30193-210, 2016 May 24.
Article de Anglais | MEDLINE | ID: mdl-27070086

RÉSUMÉ

Human mesenchymal stromal cells (MSC) have been shown to dampen immune response and promote tissue repair, but the underlying mechanisms are still under investigation. Herein, we demonstrate that umbilical cord-derived MSC (UC-MSC) alter the phenotype and function of monocyte-derived dendritic cells (DC) through lactate-mediated metabolic reprogramming. UC-MSC can secrete large quantities of lactate and, when present during monocyte-to-DC differentiation, induce instead the acquisition of M2-macrophage features in terms of morphology, surface markers, migratory properties and antigen presentation capacity. Microarray expression profiling indicates that UC-MSC modify the expression of metabolic-related genes and induce a M2-macrophage expression signature. Importantly, monocyte-derived DC obtained in presence of UC-MSC, polarize naïve allogeneic CD4+ T-cells into Th2 cells. Treatment of UC-MSC with an inhibitor of lactate dehydrogenase strongly decreases lactate concentration in culture supernatant and abrogates the effect on monocyte-to-DC differentiation. Metabolic analysis further revealed that UC-MSC decrease oxidative phosphorylation in differentiating monocytes while strongly increasing the spare respiratory capacity proportional to the amount of secreted lactate. Because both MSC and monocytes are recruited in vivo at the site of tissue damage and inflammation, we propose the local increase of lactate concentration induced by UC-MSC and the consequent enrichment in M2-macrophage generation as a mechanism to achieve immunomodulation.


Sujet(s)
Différenciation cellulaire/génétique , Acide lactique/métabolisme , Macrophages/métabolisme , Cellules souches mésenchymateuses/métabolisme , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cellules dendritiques/cytologie , Cellules dendritiques/métabolisme , Analyse de profil d'expression de gènes/méthodes , Facteur de stimulation des colonies de granulocytes et de macrophages/pharmacologie , Humains , Interleukine-4/pharmacologie , Macrophages/cytologie , Souris de lignée C57BL , Souris SCID , Monocytes/cytologie , Monocytes/métabolisme , Cordon ombilical/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...