Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
Nature ; 566(7742): 73-78, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30728521

RESUMEN

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.


Asunto(s)
Senescencia Celular/genética , Inflamación/genética , Interferón Tipo I/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Envejecimiento/genética , Envejecimiento/patología , Animales , Regulación hacia Abajo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Inflamación/patología , Lamivudine/farmacología , Masculino , Ratones , Fenotipo , ADN Polimerasa Dirigida por ARN/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología
3.
J Immunol ; 203(10): 2735-2745, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31578272

RESUMEN

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.


Asunto(s)
Linfocitos Infiltrantes de Tumor/inmunología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Traslado Adoptivo , Animales , Línea Celular Transformada/trasplante , Fibroblastos/trasplante , Vectores Genéticos , Rechazo de Injerto/inmunología , Xenoinjertos , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Quimera por Radiación , Organismos Libres de Patógenos Específicos , Telomerasa/genética , Telomerasa/fisiología , Timo/trasplante , Gelatina de Wharton/citología
4.
Blood ; 119(3): 717-26, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22101896

RESUMEN

Alterations of the BM microenvironment have been shown to occur after chemoradiotherapy, during aging, and after genetic manipulations of telomere length. Nevertheless, whether BM stromal cells adopt senescent features in response to these events is unknown. In the present study, we provide evidence that exposure to ionizing radiation (IR) leads murine stromal BM cells to express senescence markers, namely senescence-associated ß-galactosidase and increased p16(INK4a)/p19(ARF) expression. Long (8 weeks) after exposure of mice to IR, we observed a reduction in the number of stromal cells derived from BM aspirates, an effect that we found to be absent in irradiated Ink4a/arf-knockout mice and to be mostly independent of the CFU potential of the stroma. Such a reduction in the number of BM stromal cells was specific, because stromal cells isolated from collagenase-treated bones were not reduced after IR. Surprisingly, we found that exposure to IR leads to a cellular nonautonomous and Ink4a/arf-dependent effect on lymphopoiesis. Overall, our results reveal the distinct sensitivity of BM stromal cell populations to IR and suggest that long-term residual damage to the BM microenvironment can influence hematopoiesis in an Ink4a/arf-dependent manner.


Asunto(s)
Factor 1 de Ribosilacion-ADP/fisiología , Médula Ósea/efectos de la radiación , Senescencia Celular/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Homeostasis/efectos de la radiación , Radiación Ionizante , Células del Estroma/efectos de la radiación , Animales , Apoptosis , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Femenino , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Linfopoyesis/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Células del Estroma/metabolismo , Células del Estroma/patología
5.
Cytotherapy ; 16(8): 1073-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24934305

RESUMEN

BACKGROUND AIMS: Decreased bone formation with age is believed to arise, at least in part, because of the influence of the senescent microenvironment. In this context, it is unclear whether multipotent stromal cell (MSC)-based therapies would be effective for the treatment of bone diseases. METHODS: With the use of a heterotopic bone formation model, we investigated whether MSC-derived osteogenesis is impaired in aged mice compared with young mice. RESULTS: We found that bone formation derived from MSCs is not reduced in aged mice. These results are supported by the unexpected finding that conditioned media collected from ionizing radiation-induced senescent MSCs can stimulate mineralization and delay osteoclastogenesis in vitro. CONCLUSIONS: Overall, our results suggest that impaired bone formation with age is mainly cell-autonomous and provide a rationale for the use of MSC-based therapies for the treatment of bone diseases in the elderly.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Osteogénesis , Anciano , Envejecimiento/patología , Animales , Células de la Médula Ósea , Diferenciación Celular/genética , Medios de Cultivo Condicionados , Humanos , Ratones
6.
Nat Commun ; 15(1): 2435, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38499573

RESUMEN

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.


Asunto(s)
Neoplasias , Microambiente Tumoral , Animales , Ratones , Inmunoterapia , Neoplasias/patología , Senescencia Celular
7.
Cells ; 13(10)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38786071

RESUMEN

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.


Asunto(s)
Senescencia Celular , Fibroblastos , Células Madre Pluripotentes Inducidas , Humanos , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Reparación del ADN , Roturas del ADN de Doble Cadena , Estrés Fisiológico , Reprogramación Celular , Radiación Ionizante
8.
J Cell Sci ; 124(Pt 1): 68-81, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21118958

RESUMEN

DNA damage can induce a tumor suppressive response termed cellular senescence. Damaged senescent cells permanently arrest growth, secrete inflammatory cytokines and other proteins and harbor persistent nuclear foci that contain DNA damage response (DDR) proteins. To understand how persistent damage foci differ from transient foci that mark repairable DNA lesions, we identify sequential events that differentiate transient foci from persistent foci, which we term 'DNA segments with chromatin alterations reinforcing senescence' (DNA-SCARS). Unlike transient foci, DNA-SCARS associate with PML nuclear bodies, lack the DNA repair proteins RPA and RAD51, lack single-stranded DNA and DNA synthesis and accumulate activated forms of the DDR mediators CHK2 and p53. DNA-SCARS form independently of p53, pRB and several other checkpoint and repair proteins but require p53 and pRb to trigger the senescence growth arrest. Importantly, depletion of the DNA-SCARS-stabilizing component histone H2AX did not deplete 53BP1 from DNA-SCARS but diminished the presence of MDC1 and activated CHK2. Furthermore, depletion of H2AX reduced both the p53-dependent senescence growth arrest and p53-independent cytokine secretion. DNA-SCARS were also observed following severe damage to multiple human cell types and mouse tissues, suggesting that they can be used in combination with other markers to identify senescent cells. Thus, DNA-SCARS are dynamically formed distinct structures that functionally regulate multiple aspects of the senescent phenotype.


Asunto(s)
Ciclo Celular/efectos de la radiación , Núcleo Celular/efectos de la radiación , Senescencia Celular/efectos de la radiación , Cromatina/metabolismo , Citocinas/metabolismo , Daño del ADN/efectos de la radiación , Animales , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Citocinas/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Rayos X
9.
Blood ; 117(22): 6024-35, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21355092

RESUMEN

The failure of blood vessels to revascularize ischemic neural tissue represents a significant challenge for vascular biology. Examples include proliferative retinopathies (PRs) such as retinopathy of prematurity and proliferative diabetic retinopathy, which are the leading causes of blindness in children and working-age adults. PRs are characterized by initial microvascular degeneration, followed by a compensatory albeit pathologic hypervascularization mounted by the hypoxic retina attempting to reinstate metabolic equilibrium. Paradoxically, this secondary revascularization fails to grow into the most ischemic regions of the retina. Instead, the new vessels are misdirected toward the vitreous, suggesting that vasorepulsive forces operate in the avascular hypoxic retina. In the present study, we demonstrate that the neuronal guidance cue semaphorin 3A (Sema3A) is secreted by hypoxic neurons in the avascular retina in response to the proinflammatory cytokine IL-1ß. Sema3A contributes to vascular decay and later forms a chemical barrier that repels neo-vessels toward the vitreous. Conversely, silencing Sema3A expression enhances normal vascular regeneration within the ischemic retina, thereby diminishing aberrant neovascularization and preserving neuroretinal function. Overcoming the chemical barrier (Sema3A) released by ischemic neurons accelerates the vascular regeneration of neural tissues, which restores metabolic supply and improves retinal function. Our findings may be applicable to other neurovascular ischemic conditions such as stroke.


Asunto(s)
Isquemia/patología , Neovascularización Patológica , Neuronas/patología , Oxígeno/toxicidad , Regeneración , Enfermedades de la Retina/patología , Semaforina-3A/fisiología , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/metabolismo , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Técnicas para Inmunoenzimas , Interleucina-1beta/farmacología , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , ARN Mensajero/genética , Ratas , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Neovascularización Retiniana , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Nat Commun ; 14(1): 4033, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468473

RESUMEN

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.


Asunto(s)
Distrofia Miotónica , Células Satélite del Músculo Esquelético , Humanos , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Senoterapéuticos , Fibras Musculares Esqueléticas/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Desarrollo de Músculos/genética
11.
Pharmacogenomics ; 23(7): 415-430, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35485735

RESUMEN

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.


Asunto(s)
Neoplasias Pancreáticas , Leucemia-Linfoma Linfoblástico de Células Precursoras , Alelos , Asparaginasa/genética , Asparaginasa/farmacología , Asparaginasa/uso terapéutico , Proteínas de Unión al ADN/genética , Humanos , Neoplasias Pancreáticas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Neoplasias Pancreáticas
12.
Cell Rep Methods ; 2(1): 100153, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35474871

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , Neoplasias , Ratones , Humanos , Animales , Células Madre Pluripotentes Inducidas/metabolismo , Receptor de Muerte Celular Programada 1 , Neoplasias/terapia , Nivolumab , Inmunoterapia/métodos
13.
Cell Rep ; 40(7): 111241, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35977509

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , Neoplasias , Células Madre Pluripotentes , Animales , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Ratones , Neoplasias/metabolismo , Péptidos/metabolismo , Células Madre Pluripotentes/metabolismo
14.
Nature ; 435(7042): 646-51, 2005 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-15806097

RESUMEN

Permanent modification of the human genome in vivo is impractical owing to the low frequency of homologous recombination in human cells, a fact that hampers biomedical research and progress towards safe and effective gene therapy. Here we report a general solution using two fundamental biological processes: DNA recognition by C2H2 zinc-finger proteins and homology-directed repair of DNA double-strand breaks. Zinc-finger proteins engineered to recognize a unique chromosomal site can be fused to a nuclease domain, and a double-strand break induced by the resulting zinc-finger nuclease can create specific sequence alterations by stimulating homologous recombination between the chromosome and an extrachromosomal DNA donor. We show that zinc-finger nucleases designed against an X-linked severe combined immune deficiency (SCID) mutation in the IL2Rgamma gene yielded more than 18% gene-modified human cells without selection. Remarkably, about 7% of the cells acquired the desired genetic modification on both X chromosomes, with cell genotype accurately reflected at the messenger RNA and protein levels. We observe comparably high frequencies in human T cells, raising the possibility of strategies based on zinc-finger nucleases for the treatment of disease.


Asunto(s)
ADN/metabolismo , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/metabolismo , Marcación de Gen/métodos , Receptores de Interleucina-2/genética , Inmunodeficiencia Combinada Grave/genética , Dedos de Zinc , Alelos , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Células Cultivadas , Cromosomas Humanos X/genética , ADN/genética , Daño del ADN/genética , Reparación del ADN/genética , Genes Reporteros/genética , Ligamiento Genético/genética , Terapia Genética/métodos , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-2/metabolismo , Recombinación Genética/genética , Homología de Secuencia de Ácido Nucleico , Inmunodeficiencia Combinada Grave/terapia , Especificidad por Sustrato
15.
Cells ; 10(7)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206425

RESUMEN

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.


Asunto(s)
Envejecimiento/patología , Senescencia Celular , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Humanos , Modelos Biológicos , Resultado del Tratamiento
16.
Radiat Res ; 196(3): 315-322, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34107047

RESUMEN

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.


Asunto(s)
Factor de Crecimiento Epidérmico/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Neurogénesis , Protectores contra Radiación/uso terapéutico , Animales , Irradiación Craneana/efectos adversos , Proteína Doblecortina , Factor de Crecimiento Epidérmico/administración & dosificación , Factor de Crecimiento Epidérmico/genética , Femenino , Vectores Genéticos/genética , Inyecciones Intraventriculares , Lentivirus/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Protectores contra Radiación/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Transducción Genética
17.
Stem Cells Transl Med ; 10(2): 267-277, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32881406

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , Traslado Adoptivo , Animales , Diferenciación Celular , Reprogramación Celular , Fibroblastos , Trasplante de Células Madre Hematopoyéticas , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Leucocitos Mononucleares , Ratones , Mioblastos , Timo/citología
18.
Cell Metab ; 33(4): 818-832.e7, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33548171

RESUMEN

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.


Asunto(s)
Senescencia Celular , Enfermedades de la Retina/patología , Proteína bcl-X/metabolismo , Animales , Apoptosis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Cadena alfa 1 del Colágeno Tipo I/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Flavonoles/química , Flavonoles/farmacología , Flavonoles/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/metabolismo , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Proteína bcl-X/antagonistas & inhibidores
19.
J Cell Mol Med ; 14(6B): 1594-604, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19725920

RESUMEN

Patients treated for cancer therapy using ionizing radiation (IR) have delayed tissue repair and regeneration. The mechanisms mediating these defects remain largely unknown at present, thus limiting the development of therapeutic approaches. Using a wound healing model, we here investigate the mechanisms by which IR exposure limits skin regeneration. Our data show that induction of the stromal cell-derived growth factor 1alpha (SDF-1alpha) is severely impaired in the wounded skin of irradiated, compared to non-irradiated, mice. Hence, we evaluated the potential of bone marrow-derived multipotent stromal cells (MSCs), which secrete high levels of SDF-1alpha, to improve skin regeneration in irradiated mice. Injection of MSCs into the wound margin led to remarkable enhancement of skin healing in mice exposed to IR. Injection of irradiated MSCs into the wound periphery of non-irradiated mice delayed wound closure, also suggesting an important role for the stromal microenvironment in skin repair. The beneficial actions of MSCs were mainly paracrine, as the cells did not differentiate into keratinocytes. Specific knockdown of SDF-1alpha expression led to drastically reduced efficiency of MSCs in improving wound closure, indicating that SDF-1alpha secretion by MSCs is largely responsible for their beneficial action. We also found that one mechanism by which SDF-1alpha enhances wound closure likely involves increased skin vascularization. Our findings collectively indicate that SDF-1alpha is an important deregulated cytokine in irradiated wounded skin, and that the decline in tissue regeneration potential following IR can be reversed, given adequate microenvironmental support.


Asunto(s)
Células de la Médula Ósea/citología , Quimiocina CXCL12/metabolismo , Piel/patología , Piel/efectos de la radiación , Cicatrización de Heridas/efectos de la radiación , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/trasplante , Neovascularización Fisiológica/efectos de la radiación , Radiación Ionizante , Células del Estroma/metabolismo , Células del Estroma/efectos de la radiación , Células del Estroma/trasplante , Factores de Tiempo
20.
Cytotherapy ; 12(3): 394-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20331411

RESUMEN

BACKGROUND AIMS: Gene-modified mesenchymal stromal cells (MSC) provide a promising tool for cell and gene therapy-based applications by potentially acting as a cellular vehicle for protein-replacement therapy. However, to avoid the risk of insertional mutagenesis, targeted integration of a transgene into a 'safe harbor' locus is of great interest. METHODS: We sought to determine whether zinc finger nuclease (ZFN)-mediated targeted addition of the erythropoietin (Epo) gene into the chemokine [C-C motif] receptor 5 (CCR5) gene locus, a putative safe harbor locus, in MSC would result in stable transgene expression in vivo. RESULTS: Whether derived from bone marrow (BM), umbilical cord blood (UCB) or adipose tissue (AT), 30-40% of human MSC underwent ZFN-driven targeted gene addition, as determined by a combination of fluorescence-activated cell sorting (FACS)- and polymerase chain reaction (PCR)-based analyzes. An enzyme-linked immunosorbent assay (ELISA)-based analysis of gene-targeted MSC expressing Epo from the CCR5 locus showed that these modified MSC were found to secrete a significant level of Epo (c. 2 IU/10(6)cells/24 h). NOD/SCID/gammaC mice injected with ZFN-modified MSC expressing Epo exhibited significantly higher hematocrit and Epo plasma levels for several weeks post-injection, compared with mice receiving control MSC. CONCLUSIONS: These data demonstrate that MSC modified by ZFN-driven targeted gene addition may represent a cellular vehicle for delivery of plasma-soluble therapeutic factors.


Asunto(s)
Técnicas de Transferencia de Gen , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Células del Estroma/fisiología , Animales , Eritropoyetina/genética , Eritropoyetina/metabolismo , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores CCR5/genética , Células del Estroma/citología , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA