Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Gene Ther ; 29(9): 536-543, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35194185

RESUMEN

With an increasing number of gene therapy clinical trials and drugs reaching the market, it becomes important to standardize the methods that evaluate the efficacy and safety of gene therapy. We herein report the generation of lentiviral standards which are stable, cloned human cells prepared from the diploid HCT116 cell line and which carry a known number of lentiviral vector copies in their genome. These clones can be used as reference cellular materials for the calibration or qualification of analytical methods that quantify vector copy numbers in cells (VCN) or lentiviral vector genomic integration sites (IS). Cellular standards were used to show the superior precision of digital droplet PCR (ddPCR) over quantitative PCR (qPCR) for VCN determination. This enabled us to develop a new sensitive and specific VCN ddPCR method specific for the integrated provirus and not recognizing the transfer plasmid. The cellular standards, were also useful to assess the sensitivity and limits of a ligation-mediated PCR (LM-PCR) method to measure IS showing that at least 1% abundance of a single IS can be detected in a polyclonal population but that not all IS can be amplified with similar efficiency. Thus, lentiviral standards should be systematically used in all assays that assess lentiviral gene therapy efficacy and safety.


Asunto(s)
Variaciones en el Número de Copia de ADN , Terapia Genética , Genómica , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
Kidney Int ; 96(2): 350-362, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30928021

RESUMEN

Inflammation is involved in the pathogenesis of many disorders. However, the underlying mechanisms are often unknown. Here, we test whether cystinosin, the protein involved in cystinosis, is a critical regulator of galectin-3, a member of the ß-galactosidase binding protein family, during inflammation. Cystinosis is a lysosomal storage disorder and, despite ubiquitous expression of cystinosin, the kidney is the primary organ impacted by the disease. Cystinosin was found to enhance lysosomal localization and degradation of galectin-3. In Ctns-/- mice, a mouse model of cystinosis, galectin-3 is overexpressed in the kidney. The absence of galectin-3 in cystinotic mice ameliorates pathologic renal function and structure and decreases macrophage/monocyte infiltration in the kidney of the Ctns-/-Gal3-/- mice compared to Ctns-/- mice. These data strongly suggest that galectin-3 mediates inflammation involved in kidney disease progression in cystinosis. Furthermore, galectin-3 was found to interact with the pro-inflammatory cytokine Monocyte Chemoattractant Protein-1, which stimulates the recruitment of monocytes/macrophages, and proved to be significantly increased in the serum of Ctns-/- mice and also patients with cystinosis. Thus, our findings highlight a new role for cystinosin and galectin-3 interaction in inflammation and provide an additional mechanistic explanation for the kidney disease of cystinosis. This may lead to the identification of new drug targets to delay cystinosis progression.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cistinosis/complicaciones , Síndrome de Fanconi/inmunología , Galectina 3/metabolismo , Inflamación/inmunología , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Cistina/metabolismo , Cistinosis/inmunología , Cistinosis/metabolismo , Cistinosis/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Síndrome de Fanconi/metabolismo , Síndrome de Fanconi/patología , Femenino , Galectina 3/genética , Humanos , Inflamación/metabolismo , Inflamación/patología , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/patología , Lisosomas/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Monocitos/inmunología , Proteolisis
3.
Pediatr Nephrol ; 34(6): 965-973, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29789935

RESUMEN

Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders (LSDs). Initial symptoms of cystinosis correspond to the renal Fanconi syndrome. Patients then develop chronic kidney disease and multi-organ failure due to accumulation of cystine in all tissue compartments. LSDs are commonly characterized by a defective activity of lysosomal enzymes. Hematopoietic stem and progenitor cell (HSPC) transplantation is a treatment option for several LSDs based on the premise that their progeny will integrate in the affected tissues and secrete the functional enzyme, which will be recaptured by the surrounding deficient cells and restore physiological activity. However, in the case of cystinosis, the defective protein is a transmembrane lysosomal protein, cystinosin. Thus, cystinosin cannot be secreted, and yet, we showed that HSPC transplantation can rescue disease phenotype in the mouse model of cystinosis. In this review, we are describing a different mechanism by which HSPC-derived cells provide cystinosin to diseased cells within tissues, and how HSPC transplantation could be an effective one-time treatment to treat cystinosis but also other LSDs associated with a lysosomal transmembrane protein dysfunction.


Asunto(s)
Cistinosis/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Humanos
4.
Stem Cells ; 33(1): 301-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25186209

RESUMEN

Despite controversies on the potential of hematopoietic stem cells (HSCs) to promote tissue repair, we previously showed that HSC transplantation could correct cystinosis, a multisystemic lysosomal storage disease, caused by a defective lysosomal membrane cystine transporter, cystinosin (CTNS gene). Addressing the cellular mechanisms, we here report vesicular cross-correction after HSC differentiation into macrophages. Upon coculture with cystinotic fibroblasts, macrophages produced tunneling nanotubes (TNTs) allowing transfer of cystinosin-bearing lysosomes into Ctns-deficient cells, which exploited the same route to retrogradely transfer cystine-loaded lysosomes to macrophages, providing a bidirectional correction mechanism. TNT formation was enhanced by contact with diseased cells. In vivo, HSCs grafted to cystinotic kidneys also generated nanotubular extensions resembling invadopodia that crossed the dense basement membranes and delivered cystinosin into diseased proximal tubular cells. This is the first report of correction of a genetic lysosomal defect by bidirectional vesicular exchange via TNTs and suggests broader potential for HSC transplantation for other disorders due to defective vesicular proteins.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Lisosomas/metabolismo , Macrófagos/citología , Nanotubos , Animales , Cistinosis/metabolismo , Cistinosis/patología , Cistinosis/terapia , Fibroblastos , Células Madre Hematopoyéticas/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
Mol Ther ; 21(2): 433-44, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23089735

RESUMEN

Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders (LSDs). The defective gene is CTNS encoding the lysosomal cystine transporter, cystinosin. Cystine accumulates in all tissues and leads to organ damage including end-stage renal disease. Using the Ctns(-/-) murine model for cystinosis, we tested the use of hematopoietic stem and progenitor cells (HSPC) genetically modified to express a functional CTNS transgene using a self-inactivating-lentiviral vector (SIN-LV). We showed that transduced cells were capable of decreasing cystine content in all tissues and improved kidney function. Transduced HSPC retained their differentiative capabilities, populating all tissue compartments examined and allowing long-term expression of the transgene. Direct correlation between the levels of lentiviral DNA present in the peripheral blood and the levels present in tissues were demonstrated, which could be useful to follow future patients. Using a new model of cystinosis, the DsRed Ctns(-/-) mice, and a LV driving the expression of the fusion protein cystinosin-enhanced green fluorescent protein (eGFP), we showed that cystinosin was transferred from CTNS-expressing cells to Ctns-deficient adjacent cells in vitro and in vivo. This transfer led to cystine decreases in Ctns-deficient cells in vitro. These data suggest that the mechanism of cross-correction is possible in cystinosis.


Asunto(s)
Cistinosis/fisiopatología , Cistinosis/terapia , Terapia Genética/métodos , Células Madre Hematopoyéticas/citología , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Western Blotting , Cistina/análisis , Cistina/metabolismo , Cistinosis/genética , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Fallo Renal Crónico/genética , Fallo Renal Crónico/patología , Lentivirus/genética , Proteínas Luminiscentes/metabolismo , Lisosomas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética , Transgenes
6.
Biochem J ; 437(1): 63-73, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21470188

RESUMEN

Numerous anticancer agents and environmental mutagens target DNA. Although all such compounds interfere with the progression of the replication fork and inhibit DNA synthesis, there are marked differences in the DNA-damage response pathways they trigger, and the relative impact of the proximal or the distal signal transducers on cell survival is mainly lesion-specific. Accordingly, checkpoint kinase inhibitors in current clinical development show synergistic activity with some DNA-targeting agents, but not with others. In the present study, we characterize the DNA-damage response to the antitumour acronycine derivative S23906, which forms monofunctional adducts with guanine residues in the minor groove of DNA. S23906 exposure is accompanied by specific recruitment of RPA (replication protein A) at replication sites and rapid Chk1 activation. In contrast, neither MRN (Mre11-Rad50-Nbs1) nor ATM (ataxia-telangiectasia mutated), contributes to the initial response to S23906. Interestingly, genetic attenuation of ATR (ATM- and Ras3-related) activity inhibits not only the early phosphorylation of histone H2AX and Chk1, but also interferes with the late phosphorylation of Chk2. Moreover, loss of ATR function or pharmacological inhibition of the checkpoint kinases by AZD7762 is accompanied by abrogation of the S-phase arrest and increased sensitivity towards S23906. These findings identify ATR as a central co-ordinator of the DNA-damage response to S23906, and provide a mechanistic rationale for combinations of S23906 and similar agents with checkpoint abrogators.


Asunto(s)
Acronina/análogos & derivados , Antineoplásicos Alquilantes/farmacología , Proteínas de Ciclo Celular/fisiología , Daño del ADN , Mutación , Proteínas Serina-Treonina Quinasas/fisiología , Ácido Anhídrido Hidrolasas , Acronina/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Proteína Homóloga de MRE11 , Microscopía Fluorescente , Proteínas Nucleares/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Replicación A/metabolismo , Tiofenos/farmacología , Urea/análogos & derivados , Urea/farmacología
7.
Front Genome Ed ; 4: 997142, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36698790

RESUMEN

Lessons learned from decades-long practice in the transplantation of hematopoietic stem and progenitor cells (HSPCs) to treat severe inherited disorders or cancer, have set the stage for the current ex vivo gene therapies using autologous gene-modified hematopoietic stem and progenitor cells that have treated so far, hundreds of patients with monogenic disorders. With increased knowledge of hematopoietic stem and progenitor cell biology, improved modalities for patient conditioning and with the emergence of new gene editing technologies, a new era of hematopoietic stem and progenitor cell-based gene therapies is poised to emerge. Gene editing has the potential to restore physiological expression of a mutated gene, or to insert a functional gene in a precise locus with reduced off-target activity and toxicity. Advances in patient conditioning has reduced treatment toxicities and may improve the engraftment of gene-modified cells and specific progeny. Thanks to these improvements, new potential treatments of various blood- or immune disorders as well as other inherited diseases will continue to emerge. In the present review, the most recent advances in hematopoietic stem and progenitor cell gene editing will be reported, with a focus on how this approach could be a promising solution to treat non-blood-related inherited disorders and the mechanisms behind the therapeutic actions discussed.

8.
Mol Ther Methods Clin Dev ; 17: 1026-1036, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32462051

RESUMEN

Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by expansion of GAA repeats in intron 1 of the frataxin (FXN) gene, leading to significant decreased expression of frataxin, a mitochondrial iron-binding protein. We previously reported that syngeneic hematopoietic stem and progenitor cell (HSPC) transplantation prevented neurodegeneration in the FRDA mouse model YG8R. We showed that the mechanism of rescue was mediated by the transfer of the functional frataxin from HSPC-derived microglia/macrophage cells to neurons/myocytes. In this study, we report the first step toward an autologous HSPC transplantation using the CRISPR-Cas9 system for FRDA. We first identified a pair of CRISPR RNAs (crRNAs) that efficiently removes the GAA expansions in human FRDA lymphoblasts, restoring the non-pathologic level of frataxin expression and normalizing mitochondrial activity. We also optimized the gene-editing approach in HSPCs isolated from healthy and FRDA patients' peripheral blood and demonstrated normal hematopoiesis of gene-edited cells in vitro and in vivo. The procedure did not induce cellular toxic effect or major off-target events, but a p53-mediated cell proliferation delay was observed in the gene-edited cells. This study provides the foundation for the clinical translation of autologous transplantation of gene-corrected HSPCs for FRDA.

9.
Sci Transl Med ; 9(413)2017 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-29070698

RESUMEN

Friedreich's ataxia (FRDA) is an incurable autosomal recessive neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin due to an intronic GAA-repeat expansion in the FXN gene. We report the therapeutic efficacy of transplanting wild-type mouse hematopoietic stem and progenitor cells (HSPCs) into the YG8R mouse model of FRDA. In the HSPC-transplanted YG8R mice, development of muscle weakness and locomotor deficits was abrogated as was degeneration of large sensory neurons in the dorsal root ganglia (DRGs) and mitochondrial capacity was improved in brain, skeletal muscle, and heart. Transplanted HSPCs engrafted and then differentiated into microglia in the brain and spinal cord and into macrophages in the DRGs, heart, and muscle of YG8R FRDA mice. We observed the transfer of wild-type frataxin and Cox8 mitochondrial proteins from HSPC-derived microglia/macrophages to FRDA mouse neurons and muscle myocytes in vivo. Our results show the HSPC-mediated phenotypic rescue of FRDA in YG8R mice and suggest that this approach should be investigated further as a strategy for treating FRDA.


Asunto(s)
Ataxia de Friedreich/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Animales , Conducta Animal , Diferenciación Celular , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Ataxia de Friedreich/patología , Ataxia de Friedreich/fisiopatología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Unión a Hierro/metabolismo , Locomoción , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Sistema Nervioso/patología , Fagocitosis , Células Receptoras Sensoriales/patología , Frataxina
10.
Cell Cycle ; 14(13): 2080-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25945522

RESUMEN

Repair of DNA-targeted anticancer agents is an active area of investigation of both fundamental and clinical interest. However, most studies have focused on a small number of compounds limiting our understanding of both DNA repair and the DNA damage response. S23906 is an acronycine derivative that shows strong activity toward solid tumors in experimental models. S23906 forms bulky monofunctional DNA adducts in the minor groove which leads to destabilization of the double-stranded helix. We now report that S23906 induces formation of DNA double strand breaks that are processed through homologous recombination (HR) but not Non-Homologous End-Joining (NHEJ) repair. Interestingly, S23906 exposure was accompanied by a higher sensitivity of BRCA2-deficient cells compared to other HR deficient cell lines and by an S-phase accumulation in wild-type (wt), but not in BRCA2-deficient cells. Recently, we have shown that S23906-induced S phase arrest was mediated by the checkpoint kinase Chk1. However, its activated phosphorylated form is equally induced by S23906 in wt and BRCA2-deficient cells, likely indicating a role for BRCA2 downstream of Chk1. Accordingly, override of the S phase arrest by either 7-hydroxystaurosporine (UCN-01) or AZD7762 potentiates the cytotoxic activity of S23906 in wt, but not in BRCA2-deficient cells. Together, our findings suggest that the pronounced sensitivity of BRCA2-deficient cells to S23906 is due to both a defective S-phase arrest and the absence of HR repair. Tumors with deficiencies for proteins involved in HR, and BRCA2 in particular, may thus show increased sensitivity to S23906, thereby providing a rationale for patient selection in clinical trials.


Asunto(s)
Acronina/análogos & derivados , Antineoplásicos Alquilantes/metabolismo , Proteína BRCA2/metabolismo , Puntos de Control del Ciclo Celular/fisiología , Reparación del ADN/fisiología , Acronina/metabolismo , Acronina/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cricetinae , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/fisiología , Reparación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos
11.
Invest Ophthalmol Vis Sci ; 56(12): 7214-23, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26540660

RESUMEN

PURPOSE: Cystinosis is caused by a deficiency in the lysosomal cystine transporter, cystinosin (CTNS gene), resulting in cystine crystal accumulation in tissues. In eyes, crystals accumulate in the cornea causing photophobia and eventually blindness. Hematopoietic stem progenitor cells (HSPCs) rescue the kidney in a mouse model of cystinosis. We investigated the potential for HSPC transplantation to treat corneal defects in cystinosis. METHODS: We isolated HSPCs from transgenic DsRed mice and systemically transplanted irradiated Ctns-/- mice. A year posttransplantation, we investigated the fate and function of HSPCs by in vivo confocal and fluorescence microscopy (IVCM), quantitative RT-PCR (RT-qPCR), mass spectrometry, histology, and by measuring the IOP. To determine the mechanism by which HSPCs may rescue disease cells, we transplanted Ctns-/- mice with Ctns-/- DsRed HSPCs virally transduced to express functional CTNS-eGFP fusion protein. RESULTS: We found that a single systemic transplantation of wild-type HSPCs prevented ocular pathology in the Ctns-/- mice. Engraftment-derived HSPCs were detected within the cornea, and also in the sclera, ciliary body, retina, choroid, and lens. Transplantation of HSPC led to substantial decreases in corneal cystine crystals, restoration of normal corneal thickness, and lowered IOP in mice with high levels of donor-derived cell engraftment. Finally, we found that HSPC-derived progeny differentiated into macrophages, which displayed tunneling nanotubes capable of transferring cystinosin-bearing lysosomes to diseased cells. CONCLUSIONS: To our knowledge, this is the first demonstration that HSPCs can rescue hereditary corneal defects, and supports a new potential therapeutic strategy for treating ocular pathologies.


Asunto(s)
Cistinosis/terapia , Oftalmopatías/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Células Cultivadas , Cistinosis/genética , Modelos Animales de Enfermedad , Oftalmopatías/congénito , Oftalmopatías/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
12.
EMBO Mol Med ; 7(2): 158-74, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25586965

RESUMEN

Metabolite accumulation in lysosomal storage disorders (LSDs) results in impaired cell function and multi-systemic disease. Although substrate reduction and lysosomal overload-decreasing therapies can ameliorate disease progression, the significance of lysosomal overload-independent mechanisms in the development of cellular dysfunction is unknown for most LSDs. Here, we identify a mechanism of impaired chaperone-mediated autophagy (CMA) in cystinosis, a LSD caused by defects in the cystine transporter cystinosin (CTNS) and characterized by cystine lysosomal accumulation. We show that, different from other LSDs, autophagosome number is increased, but macroautophagic flux is not impaired in cystinosis while mTOR activity is not affected. Conversely, the expression and localization of the CMA receptor LAMP2A are abnormal in CTNS-deficient cells and degradation of the CMA substrate GAPDH is defective in Ctns(-/-) mice. Importantly, cysteamine treatment, despite decreasing lysosomal overload, did not correct defective CMA in Ctns(-/-) mice or LAMP2A mislocalization in cystinotic cells, which was rescued by CTNS expression instead, suggesting that cystinosin is important for CMA activity. In conclusion, CMA impairment contributes to cell malfunction in cystinosis, highlighting the need for treatments complementary to current therapies that are based on decreasing lysosomal overload.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Autofagia , Cistinosis/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Chaperonas Moleculares/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Cistina/metabolismo , Cistinosis/genética , Cistinosis/fisiopatología , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética
13.
Methods Mol Biol ; 1114: 325-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24557913

RESUMEN

Gene targeting by homologous recombination at chromosomal endogenous loci has traditionally been considered a low-efficiency process. However, the effectiveness of such so-called genome surgery or genome editing has recently been drastically improved through technical developments, chiefly the use of designer nucleases like zinc-finger nucleases (ZFNs), meganucleases, transcription activator-like effector nucleases (TALENs) and CRISPR/Cas nucleases. These enzymes are custom designed to recognize long target sites and introduce double-strand breaks (DSBs) at specific target loci in the genome, which in turn mediate significant improvements in the frequency of homologous recombination. Here, we describe a Southern blot-based assay that allows detection of gene repair and estimation of repair frequencies in a cell population, useful in cases where the targeted modification itself cannot be detected by restriction digest. This is achieved through detection of a silent restriction site introduced alongside the desired mutation, in our particular example using integration-deficient lentiviral vectors (IDLVs) coding for ZFNs and a suitable DNA repair template.


Asunto(s)
Southern Blotting , Reparación del ADN , Endonucleasas/metabolismo , Dedos de Zinc/fisiología , Animales , Southern Blotting/métodos , Endonucleasas/genética , Fibroblastos/metabolismo , Vectores Genéticos/genética , Recombinación Homóloga , Lentivirus/genética , Ratones , Transducción Genética
14.
Mol Cell Biol ; 33(15): 2950-62, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23716592

RESUMEN

Cystinosis is a lysosomal storage disorder caused by the accumulation of the amino acid cystine due to genetic defects in the CTNS gene, which encodes cystinosin, the lysosomal cystine transporter. Although many cellular dysfunctions have been described in cystinosis, the mechanisms leading to these defects are not well understood. Here, we show that increased lysosomal overload induced by accumulated cystine leads to cellular abnormalities, including vesicular transport defects and increased endoplasmic reticulum (ER) stress, and that correction of lysosomal transport improves cellular function in cystinosis. We found that Rab27a was expressed in proximal tubular cells (PTCs) and partially colocalized with the lysosomal marker LAMP-1. The expression of Rab27a but not other small GTPases, including Rab3 and Rab7, was downregulated in kidneys from Ctns-/- mice and in human PTCs from cystinotic patients. Using total internal reflection fluorescence microscopy, we found that lysosomal transport is impaired in Ctns-/- cells. Ctns-/- cells showed significant ER expansion and a marked increase in the unfolded protein response-induced chaperones Grp78 and Grp94. Upregulation of the Rab27a-dependent vesicular trafficking mechanisms rescued the defective lysosomal transport phenotype and reduced ER stress in cystinotic cells. Importantly, reconstitution of lysosomal transport mediated by Rab27a led to decreased lysosomal overload, manifested as reduced cystine cellular content. Our data suggest that upregulation of the Rab27a-dependent lysosomal trafficking and secretory pathways contributes to the correction of some of the cellular defects induced by lysosomal overload in cystinosis, including ER stress.


Asunto(s)
Cistinosis/genética , Cistinosis/patología , Estrés del Retículo Endoplásmico , Lisosomas/patología , Proteínas de Unión al GTP rab/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Transporte Biológico , Calcio/metabolismo , Línea Celular , Células Cultivadas , Cistina/metabolismo , Cistinosis/metabolismo , Regulación hacia Abajo , Chaperón BiP del Retículo Endoplásmico , Exocitosis , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba , Proteínas de Unión al GTP rab/metabolismo , Proteínas rab27 de Unión a GTP
15.
Mol Cancer Ther ; 10(8): 1481-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21622731

RESUMEN

PM01183 is a novel marine-derived covalent DNA binder in clinical development. PM01183 is structurally similar to trabectedin (yondelis, ecteinascidin-743) except for the C subunit, and this modification is accompanied by different pharmacokinetics in cancer patients. We here characterize the interaction of PM01183 with the nucleotide excision repair (NER) pathway in comparison with trabectedin. Our results show for the first time that although neither PM01183 nor trabectedin is repaired by NER, both compounds are able to interfere with the NER machinery thereby attenuating the repair of specific NER substrates. We further show that the NER activity is increased in 3 of 4 cellular models with acquired resistance to cisplatin or oxaliplatin, confirming the involvement of NER in the resistance to platinum derivatives. Importantly, both PM01183 and trabectedin show unchanged or even enhanced activity toward all 4 cisplatin- and oxaliplatin-resistant cell lines. We finally show that combinations of PM01183 and cisplatin were mostly synergistic toward both parental and cisplatin-resistant ovarian carcinoma cells as indicated by Chou and Talalay analysis. These data show that the C subunit of trabectedin can be subjected to at least some structural modifications without loss of activity or NER interaction. While PM01183 and trabectedin appear functionally similar in cellular models, it is likely that the differences in pharmacokinetics may allow different dosing and scheduling of PM01183 in the clinic that could lead to novel and/or increased antitumor activity. Taken together, our results provide a mechanistic basis to support clinical trials of PM01183 alone or in combination with cisplatin.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Carbolinas/farmacología , Cisplatino/farmacología , Reparación del ADN/efectos de los fármacos , Dioxoles/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Tetrahidroisoquinolinas/farmacología , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Carbolinas/química , Línea Celular Tumoral , Cromatina/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Dioxoles/química , Dioxoles/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Células HCT116 , Células HT29 , Compuestos Heterocíclicos de 4 o más Anillos/química , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Compuestos Organoplatinos/farmacología , Oxaliplatino , Tetrahidroisoquinolinas/química , Tetrahidroisoquinolinas/metabolismo , Trabectedina , Rayos Ultravioleta/efectos adversos
16.
Biochem Pharmacol ; 80(3): 335-43, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20399198

RESUMEN

S23906 belongs to a novel class of alkylating anticancer agents forming bulky monofunctional DNA adducts. A unique feature of S23906 is its "helicase-like" activity leading to the destabilization of the surrounding duplex DNA. We here characterize the recognition and repair of S23906 adducts by the nucleotide excision repair (NER) machinery. All NER-deficient human cell lines tested showed increased sensitivity to S23906, which was particularly pronounced for cells deficient in XPC, CSB and XPA. In comparison, deficiencies in ERCC1 or XPF had lesser impact on the sensitivity to S23906. The sensitivity was, at least in part, linked to the conversion of unrepaired adducts into toxic DNA strand breaks as shown by single cell electrophoresis and gamma-H2AX formation. The pharmacological relevance of these findings was confirmed by the characterization of KB carcinoma cells with acquired S23906 resistance. These cells showed increased NER activity in vivo as well as toward damaged plasmid DNA in vitro. In particular, both global genome NER, as shown by unscheduled DNA synthesis, and transcription-coupled NER, as shown by transcriptional recovery, were up-regulated in the S23906-resistant cells. The increased NER activity was accompanied by up to 5-fold up-regulation of XPC, CSB and XPA proteins without detectable alterations of ERCC1 on the DNA, RNA or protein levels. Our results suggest that S23906 adducts are recognized and repaired by both NER sub-pathways in contrast to other members of this class, that are only recognized by transcription-coupled NER. We further show that NER activity can be up-regulated without changes in ERCC1 expression.


Asunto(s)
Acronina/farmacología , Antineoplásicos Alquilantes/metabolismo , ADN Helicasas/fisiología , Enzimas Reparadoras del ADN/fisiología , Proteínas de Unión al ADN/fisiología , ADN/metabolismo , Endonucleasas/fisiología , Acronina/metabolismo , Alquilantes/química , Alquilantes/metabolismo , Antineoplásicos Alquilantes/química , Sitios de Unión/fisiología , Línea Celular , Línea Celular Tumoral , Aductos de ADN/metabolismo , Humanos , Receptores X del Hígado , Receptores Nucleares Huérfanos/deficiencia , Receptores Nucleares Huérfanos/fisiología , Proteínas de Unión a Poli-ADP-Ribosa , Xerodermia Pigmentosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA